101
|
Xu W, Liu F, Li Q, Li L, Liu X. Integrated Analysis of miRNA and mRNA Regulation Network in Hypertension. Biochem Genet 2023; 61:2566-2579. [PMID: 37165183 DOI: 10.1007/s10528-023-10389-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/18/2023] [Indexed: 05/12/2023]
Abstract
Hypertension is the most common chronic disease. Early diagnosis is helpful for early medical intervention. The miRNAs and the messenger RNAs (mRNAs) network may be valuable disease diagnosis markers. We aimed to explore the diagnostic value of the miRNA-mRNA network for hypertension patients. Data of miRNAs and mRNAs expression were obtained from the Gene Expression Omnibus database. The weighted gene co-expression network analysis was performed to screen hypertension-related gene modules, and these genes undergone functional enrichment analysis using "clusterProfiler" package. Differential expression analysis was applied on miRNAs expression profiles using "limma" package. TargetScanHuman and miRDB databases were used to select target mRNAs. Cytoscape software was used to visualize the miRNA-mRNA regulation network. P value < 0.05 was considered statistically significant after t test. There were 123 screened mRNAs which were enriched in 161 Gene ontology (GO) terms and 14 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Thirty-five differentially expressed miRNAs (DEMs) are found in the GSE75670. Totally 36 miRNA-mRNA pairs were obtained after the integrated analysis, and three mRNAs and the hsa-miRNA-5589-5p were identified as key joints. Hub genes, KIAA0513, ARID3A and LRPAP1, and key hsa-miRNA-5589-5p are potential diagnostic biomarkers for hypertension. Our findings are promising in the clinical application, conducive to early detection and prompt intervention of hypertension.
Collapse
Affiliation(s)
- Weijuan Xu
- Department of Geriatrics, Zibo Central Hospital, No 54 Gongqingtuan West Road, 255036, Zibo, People's Republic of China
| | - Feng Liu
- Department of Emergency, Zibo Central Hospital, No 54 Gongqingtuan West Road, 255036, Zibo, People's Republic of China
| | - Qinghua Li
- Department of Emergency, Zibo Central Hospital, No 54 Gongqingtuan West Road, 255036, Zibo, People's Republic of China
| | - Li Li
- Department of Geriatrics, Zibo Central Hospital, No 54 Gongqingtuan West Road, 255036, Zibo, People's Republic of China.
| | - Xiqiang Liu
- Department of Geriatrics, Zibo Central Hospital, No 54 Gongqingtuan West Road, 255036, Zibo, People's Republic of China
| |
Collapse
|
102
|
Ferreira B, Heredia A, Serpa J. An integrative view on glucagon function and putative role in the progression of pancreatic neuroendocrine tumours (pNETs) and hepatocellular carcinomas (HCC). Mol Cell Endocrinol 2023; 578:112063. [PMID: 37678603 DOI: 10.1016/j.mce.2023.112063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/16/2023] [Accepted: 09/02/2023] [Indexed: 09/09/2023]
Abstract
Cancer metabolism research area evolved greatly, however, is still unknown the impact of systemic metabolism control and diet on cancer. It makes sense that systemic regulators of metabolism can act directly on cancer cells and activate signalling, prompting metabolic remodelling needed to sustain cancer cell survival, tumour growth and disease progression. In the present review, we describe the main glucagon functions in the control of glycaemia and of metabolic pathways overall. Furthermore, an integrative view on how glucagon and related signalling pathways can contribute for pancreatic neuroendocrine tumours (pNETs) and hepatocellular carcinomas (HCC) progression, since pancreas and liver are the major organs exposed to higher levels of glucagon, pancreas as a producer and liver as a scavenger. The main objective is to bring to discussion some glucagon-dependent mechanisms by presenting an integrative view on microenvironmental and systemic aspects in pNETs and HCC biology.
Collapse
Affiliation(s)
- Bárbara Ferreira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo Dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal
| | - Adrián Heredia
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo Dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal; Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz MB, 1649-028, Lisboa, Portugal
| | - Jacinta Serpa
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo Dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal.
| |
Collapse
|
103
|
Shen X, Niu N, Xue J. Oncogenic KRAS triggers metabolic reprogramming in pancreatic ductal adenocarcinoma. J Transl Int Med 2023; 11:322-329. [PMID: 38130635 PMCID: PMC10732496 DOI: 10.2478/jtim-2022-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with an extremely high lethality rate. Oncogenic KRAS activation has been proven to be a key driver of PDAC initiation and progression. There is increasing evidence that PDAC cells undergo extensive metabolic reprogramming to adapt to their extreme energy and biomass demands. Cell-intrinsic factors, such as KRAS mutations, are able to trigger metabolic rewriting. Here, we update recent advances in KRAS-driven metabolic reprogramming and the associated metabolic therapeutic potential in PDAC.
Collapse
Affiliation(s)
- Xuqing Shen
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai200127, China
| | - Ningning Niu
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai200127, China
| | - Jing Xue
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai200127, China
| |
Collapse
|
104
|
Qiu X, Ye H, Li X, Li D, Jiang L, Liu R, Zhao Z, He D. IL-6/JAK2-dependent G6PD phosphorylation promotes nucleotide synthesis and supports tumor growth. Mol Metab 2023; 78:101836. [PMID: 37949355 PMCID: PMC10692918 DOI: 10.1016/j.molmet.2023.101836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVE Tumor cells hijack inflammatory mechanisms to promote their own growth. IL-6 is one of the major cytokines, and is frequently upregulated in tumors. The pentose phosphate pathway (PPP) generates the indispensable building blocks to produce various nucleotides. Here we aimed to determine whether and how PPP is timely tuned in response to IL-6 to support tumor growth. METHODS Protein expression was examined by immunoblot. Protein interaction was examined by immunoprecipitation. Tumor cell proliferation in in vitro culture was examined by BrdU assay and colony formation assay. Tumor cell proliferation in mouse xenograft model was examined by Ki-67 staining. RESULTS Here we show that the metabolic flux of PPP and enzymatic activity of glucose-6-phosphate dehydrogenase (G6PD) is rapidly induced under IL-6 treatment, without obvious changes in G6PD expression level. Mechanistically, Janus kinase 2 (JAK2) phosphorylates G6PD Y437 under IL-6 treatment, which accentuates G6PD enzymatic activity by promoting G6PD binding with its substrate G6P. Further, JAK2-dependent G6PD Y437 phosphorylation is required for IL-6-induced nucleotide biosynthesis and tumor cell proliferation, and is associated with the progression of oral squamous cell carcinoma. CONCLUSIONS Our findings report a new mechanism implicated in the crosstalk between tumor cells and inflammatory microenvironment, by which JAK2-dependent activation of G6PD governs nucleotide synthesis to support tumor cell proliferation, thereby highlighting its value as a potential anti-tumor target.
Collapse
Affiliation(s)
- Xuemei Qiu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Hongping Ye
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Xiaofei Li
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, 610057, PR China
| | - Dan Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Lu Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China.
| | - Rui Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China.
| | - Zhe Zhao
- Nuclear Stress Medicine Center, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, 610057, PR China.
| | - Dan He
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, 610057, PR China.
| |
Collapse
|
105
|
Nakamizo S, Sugiura Y, Ishida Y, Ueki Y, Yonekura S, Tanizaki H, Date H, Yoshizawa A, Murata T, Minatoya K, Katagiri M, Nomura S, Komuro I, Ogawa S, Nakajima S, Kambe N, Egawa G, Kabashima K. Activation of the pentose phosphate pathway in macrophages is crucial for granuloma formation in sarcoidosis. J Clin Invest 2023; 133:e171088. [PMID: 38038136 PMCID: PMC10688990 DOI: 10.1172/jci171088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/27/2023] [Indexed: 12/02/2023] Open
Abstract
Sarcoidosis is a disease of unknown etiology in which granulomas form throughout the body and is typically treated with glucocorticoids, but there are no approved steroid-sparing alternatives. Here, we investigated the mechanism of granuloma formation using single-cell RNA-Seq in sarcoidosis patients. We observed that the percentages of triggering receptor expressed on myeloid cells 2-positive (TREM2-positive) macrophages expressing angiotensin-converting enzyme (ACE) and lysozyme, diagnostic makers of sarcoidosis, were increased in cutaneous sarcoidosis granulomas. Macrophages in the sarcoidosis lesion were hypermetabolic, especially in the pentose phosphate pathway (PPP). Expression of the PPP enzymes, such as fructose-1,6-bisphosphatase 1 (FBP1), was elevated in both systemic granuloma lesions and serum of sarcoidosis patients. Granuloma formation was attenuated by the PPP inhibitors in in vitro giant cell and in vivo murine granuloma models. These results suggest that the PPP may be a promising target for developing therapeutics for sarcoidosis.
Collapse
Affiliation(s)
- Satoshi Nakamizo
- Department of Dermatology
- Alliance Laboratory for Advanced Medical Research, and
| | - Yuki Sugiura
- Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Yoko Ueki
- Department of Dermatology, Kansai Medical University, Osaka, Japan
| | | | - Hideaki Tanizaki
- Department of Dermatology, Kansai Medical University, Osaka, Japan
| | | | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Teruasa Murata
- Department of Dermatology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Seitaro Nomura
- Department of Cardiovascular Medicine and
- Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo
| | - Issei Komuro
- Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo
- International University of Health and Welfare, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Saeko Nakajima
- Department of Dermatology
- Department of Drug Discovery for Inflammatory Skin Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | - Kenji Kabashima
- Department of Dermatology
- Skin Research Institute of Singapore (SRIS) and A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), Singapore
| |
Collapse
|
106
|
Zhang L, Deng Y, Yang J, Deng W, Li L. Neurotransmitter receptor-related gene signature as potential prognostic and therapeutic biomarkers in colorectal cancer. Front Cell Dev Biol 2023; 11:1202193. [PMID: 38099288 PMCID: PMC10720326 DOI: 10.3389/fcell.2023.1202193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
Background: Colorectal cancer is one of the most common malignant tumors worldwide. A various of neurotransmitter receptors have been found to be expressed in tumor cells, and the activation of these receptors may promote tumor growth and metastasis. This study aimed to construct a novel neurotransmitter receptor-related genes signature to predict the survival, immune microenvironment, and treatment response of colorectal cancer patients. Methods: RNA-seq and clinical data of colorectal cancer from The Cancer Genome Atlas database and Gene Expression Omnibus were downloaded. Neurotransmitter receptor-related gene were collected from publicly available data sources. The Weighted Gene Coexpression Network Analysis (WGCNA), Least Absolute Shrinkage and Selection Operator (LASSO) logistic regression, Support Vector Machine-Recursive Feature Elimination (SVM-RFE), and Random Forest (RF) algorithms were employed to construct the Neurotransmitter receptor-related gene prognostic signature. Further analyses, functional enrichment, CIBERSORTx, The Tumor Immune Single Cell Center (TISCH), survival analysis, and CellMiner, were performed to analyze immune status and treatment responses. Quantitative real-time polymerase chain reaction (qRT-PCR) assays were carried out to confirm the expression levels of prognostic genes. Results: By combining machine learning algorithm and WGCNA, we identified CHRNA3, GABRD, GRIK3, and GRIK5 as Neurotransmitter receptor-related prognostic genes signature. Functional enrichment analyses showed that these genes were enriched with cellular metabolic-related pathways, such as organic acid, inorganic acid, and lipid metabolism. CIBERSORTx and Single cell analysis showed that the high expression of genes were positively correlated with immunosuppressive cells infiltration, and the genes were mainly expressed in cancer-associated fibroblasts and endothelial cells. A nomogram was further built to predict overall survival (OS). The expression of CHRNA3, GABRD, GRIK3, and GRIK5 in cancer cells significantly impacted their response to chemotherapy. Conclusion: A neurotransmitter receptor-related prognostic gene signature was developed and validated in the current study, giving novel sights of neurotransmitter in predicting the prognostic and improving the treatment of CRC.
Collapse
Affiliation(s)
- Linjie Zhang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yizhang Deng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jingbang Yang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Wuguo Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Liren Li
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
107
|
Arumugam P, Kielian T. Metabolism Shapes Immune Responses to Staphylococcus aureus. J Innate Immun 2023; 16:12-30. [PMID: 38016430 PMCID: PMC10766399 DOI: 10.1159/000535482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Staphylococcus aureus (S. aureus) is a common cause of hospital- and community-acquired infections that can result in various clinical manifestations ranging from mild to severe disease. The bacterium utilizes different combinations of virulence factors and biofilm formation to establish a successful infection, and the emergence of methicillin- and vancomycin-resistant strains introduces additional challenges for infection management and treatment. SUMMARY Metabolic programming of immune cells regulates the balance of energy requirements for activation and dictates pro- versus anti-inflammatory function. Recent investigations into metabolic adaptations of leukocytes and S. aureus during infection indicate that metabolic crosstalk plays a crucial role in pathogenesis. Furthermore, S. aureus can modify its metabolic profile to fit an array of niches for commensal or invasive growth. KEY MESSAGES Here we focus on the current understanding of immunometabolism during S. aureus infection and explore how metabolic crosstalk between the host and S. aureus influences disease outcome. We also discuss how key metabolic pathways influence leukocyte responses to other bacterial pathogens when information for S. aureus is not available. A better understanding of how S. aureus and leukocytes adapt their metabolic profiles in distinct tissue niches may reveal novel therapeutic targets to prevent or control invasive infections.
Collapse
Affiliation(s)
- Prabhakar Arumugam
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
108
|
Koopmans PJ, Ismaeel A, Goljanek-Whysall K, Murach KA. The roles of miRNAs in adult skeletal muscle satellite cells. Free Radic Biol Med 2023; 209:228-238. [PMID: 37879420 PMCID: PMC10911817 DOI: 10.1016/j.freeradbiomed.2023.10.403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023]
Abstract
Satellite cells are bona fide muscle stem cells that are indispensable for successful post-natal muscle growth and regeneration after severe injury. These cells also participate in adult muscle adaptation in several capacities. MicroRNAs (miRNAs) are post-transcriptional regulators of mRNA that are implicated in several aspects of stem cell function. There is evidence to suggest that miRNAs affect satellite cell behavior in vivo during development and myogenic progenitor behavior in vitro, but the role of miRNAs in adult skeletal muscle satellite cells is less studied. In this review, we provide evidence for how miRNAs control satellite cell function with emphasis on satellite cells of adult skeletal muscle in vivo. We first outline how miRNAs are indispensable for satellite cell viability and control the phases of myogenesis. Next, we discuss the interplay between miRNAs and myogenic cell redox status, senescence, and communication to other muscle-resident cells during muscle adaptation. Results from recent satellite cell miRNA profiling studies are also summarized. In vitro experiments in primary myogenic cells and cell lines have been invaluable for exploring the influence of miRNAs, but we identify a need for novel genetic tools to further interrogate how miRNAs control satellite cell behavior in adult skeletal muscle in vivo.
Collapse
Affiliation(s)
- Pieter Jan Koopmans
- Exercise Science Research Center, Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA; Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Katarzyna Goljanek-Whysall
- School of Medicine, College of Medicine, Nursing, and Health Sciences, University of Galway, Galway, Ireland
| | - Kevin A Murach
- Exercise Science Research Center, Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA; Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, 72701, USA.
| |
Collapse
|
109
|
Patronas EM, Balber T, Miller A, Geist BK, Michligk A, Vraka C, Krisch M, Rohr-Udilova N, Haschemi A, Viernstein H, Hacker M, Mitterhauser M. A fingerprint of 2-[ 18F]FDG radiometabolites - How tissue-specific metabolism beyond 2-[ 18F]FDG-6-P could affect tracer accumulation. iScience 2023; 26:108137. [PMID: 37867937 PMCID: PMC10585399 DOI: 10.1016/j.isci.2023.108137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/01/2023] [Accepted: 10/02/2023] [Indexed: 10/24/2023] Open
Abstract
Studies indicate that the radiotracer 2-[18F]fluoro-2-deoxy-D-glucose (2-[18F]FDG) can be metabolized beyond 2-[18F]FDG-6-phosphate (2-[18F]FDG-6-P), but its metabolism is incompletely understood. Most importantly, it remains unclear whether downstream metabolism affects tracer accumulation in vivo. Here we present a fingerprint of 2-[18F]FDG radiometabolites over time in cancer cells, corresponding tumor xenografts and murine organs. Strikingly, radiometabolites representing glycogen metabolism or the oxPPP correlated inversely with tracer accumulation across all examined tissues. Recent studies suggest that not only hexokinase, but also hexose-6-phosphate dehydrogenase (H6PD), an enzyme of the oxidative pentose phosphate pathway (oxPPP), determines 2-[18F]FDG accumulation. However, little is known about the corresponding enzyme glucose-6-phosphate dehydrogenase (G6PD). Our mechanistic in vitro experiments on the role of the oxPPP propose that 2-[18F]FDG can be metabolized via both G6PD and H6PD, but data from separate enzyme knockdown suggest diverging roles in downstream tracer metabolism. Overall, we propose that tissue-specific metabolism beyond 2-[18F]FDG-6-P could matter for imaging.
Collapse
Affiliation(s)
- Eva-Maria Patronas
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
- Division of Pharmaceutical Technology and Biopharmaceutics, Department of Pharmaceutical Sciences, University of Vienna, Vienna 1090, Austria
| | - Theresa Balber
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna 1090, Austria
| | - Anne Miller
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna 1090, Austria
| | - Barbara Katharina Geist
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Antje Michligk
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Chrysoula Vraka
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Maximilian Krisch
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Nataliya Rohr-Udilova
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Helmut Viernstein
- Division of Pharmaceutical Technology and Biopharmaceutics, Department of Pharmaceutical Sciences, University of Vienna, Vienna 1090, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Markus Mitterhauser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna 1090, Austria
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry, Vienna 1090, Austria
| |
Collapse
|
110
|
Scheibelberger L, Stankovic T, Pühringer M, Kählig H, Balber T, Patronas E, Rampler E, Mitterhauser M, Haschemi A, Pallitsch K. Synthesis of 4-Deoxy-4-Fluoro-d-Sedoheptulose: A Promising New Sugar to Apply the Principle of Metabolic Trapping. Chemistry 2023; 29:e202302277. [PMID: 37552007 PMCID: PMC10946558 DOI: 10.1002/chem.202302277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/09/2023]
Abstract
Fluorinated carbohydrates are important tools for understanding the deregulation of metabolic fluxes and pathways. Fluorinating specific positions within the sugar scaffold can lead to enhanced metabolic stability and subsequent metabolic trapping in cells. This principle has, however, never been applied to study the metabolism of the rare sugars of the pentose phosphate pathway (PPP). In this study, two fluorinated derivatives of d-sedoheptulose were designed and synthesized: 4-deoxy-4-fluoro-d-sedoheptulose (4DFS) and 3-deoxy-3-fluoro-d-sedoheptulose (3DFS). Both sugars are taken up by human fibroblasts but only 4DFS is phosphorylated. Fluorination of d-sedoheptulose at C-4 effectively halts the enzymatic degradation by transaldolase and transketolase. 4DFS thus has a high potential as a new PPP imaging probe based on the principle of metabolic trapping. Therefore, the synthesis of potential radiolabeling precursors for 4DFS for future radiofluorinations with fluorine-18 is presented.
Collapse
Affiliation(s)
- Lukas Scheibelberger
- Institute of Organic ChemistryUniversity of ViennaWähringer Straße 381090ViennaAustria
- Vienna Doctoral School in Chemistry (DoSChem)University of ViennaWähringer Straße 421090ViennaAustria
| | - Toda Stankovic
- Institute of Organic ChemistryUniversity of ViennaWähringer Straße 381090ViennaAustria
| | - Marlene Pühringer
- Vienna Doctoral School in Chemistry (DoSChem)University of ViennaWähringer Straße 421090ViennaAustria
- Institute of Analytical ChemistryUniversity of ViennaWähringer Straße 381090ViennaAustria
| | - Hanspeter Kählig
- Institute of Organic ChemistryUniversity of ViennaWähringer Straße 381090ViennaAustria
| | - Theresa Balber
- Division of Nuclear MedicineDepartment of Biomedical Imaging and Image-guided TherapyMedical University of ViennaWähringer Gürtel 18–201090ViennaAustria
- Ludwig Boltzmann Institute Applied DiagnosticsWähringer Gürtel 18–201090ViennaAustria
| | - Eva‐Maria Patronas
- Division of Nuclear MedicineDepartment of Biomedical Imaging and Image-guided TherapyMedical University of ViennaWähringer Gürtel 18–201090ViennaAustria
- Division of Pharmaceutical Technology and BiopharmaceuticsDepartment of Pharmaceutical SciencesUniversity of Vienna, UZAIIJosef-Holaubek-Platz 21090ViennaAustria
| | - Evelyn Rampler
- Institute of Analytical ChemistryUniversity of ViennaWähringer Straße 381090ViennaAustria
| | - Markus Mitterhauser
- Division of Nuclear MedicineDepartment of Biomedical Imaging and Image-guided TherapyMedical University of ViennaWähringer Gürtel 18–201090ViennaAustria
- Ludwig Boltzmann Institute Applied DiagnosticsWähringer Gürtel 18–201090ViennaAustria
- Institute of Inorganic ChemistryUniversity of ViennaWähringer Straße 421090ViennaAustria
| | - Arvand Haschemi
- Department of Laboratory MedicineMedical University of ViennaWähringer Gürtel 18–201090ViennaAustria
| | - Katharina Pallitsch
- Institute of Organic ChemistryUniversity of ViennaWähringer Straße 381090ViennaAustria
| |
Collapse
|
111
|
Terlizzi C, De Rosa V, Iommelli F, Pezone A, Altobelli GG, Maddalena M, Dimitrov J, De Rosa C, Della Corte CM, Avvedimento VE, Del Vecchio S. ATM inhibition blocks glucose metabolism and amplifies the sensitivity of resistant lung cancer cell lines to oncogene driver inhibitors. Cancer Metab 2023; 11:20. [PMID: 37932830 PMCID: PMC10629204 DOI: 10.1186/s40170-023-00320-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 10/14/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND ATM is a multifunctional serine/threonine kinase that in addition to its well-established role in DNA repair mechanisms is involved in a number of signaling pathways including regulation of oxidative stress response and metabolic diversion of glucose through the pentose phosphate pathway. Oncogene-driven tumorigenesis often implies the metabolic switch from oxidative phosphorylation to glycolysis which provides metabolic intermediates to sustain cell proliferation. The aim of our study is to elucidate the role of ATM in the regulation of glucose metabolism in oncogene-driven cancer cells and to test whether ATM may be a suitable target for anticancer therapy. METHODS Two oncogene-driven NSCLC cell lines, namely H1975 and H1993 cells, were treated with ATM inhibitor, KU55933, alone or in combination with oncogene driver inhibitors, WZ4002 or crizotinib. Key glycolytic enzymes, mitochondrial complex subunits (OXPHOS), cyclin D1, and apoptotic markers were analyzed by Western blotting. Drug-induced toxicity was assessed by MTS assay using stand-alone or combined treatment with KU55933 and driver inhibitors. Glucose consumption, pyruvate, citrate, and succinate levels were also analyzed in response to KU55933 treatment. Both cell lines were transfected with ATM-targeted siRNA or non-targeting siRNA and then exposed to treatment with driver inhibitors. RESULTS ATM inhibition deregulates and inhibits glucose metabolism by reducing HKII, p-PKM2Tyr105, p-PKM2Ser37, E1α subunit of pyruvate dehydrogenase complex, and all subunits of mitochondrial complexes except ATP synthase. Accordingly, glucose uptake and pyruvate concentrations were reduced in response to ATM inhibition, whereas citrate and succinate levels were increased in both cell lines indicating the supply of alternative metabolic substrates. Silencing of ATM resulted in similar changes in glycolytic cascade and OXPHOS levels. Furthermore, the driver inhibitors amplified the effects of ATM downregulation on glucose metabolism, and the combined treatment with ATM inhibitors enhanced the cytotoxic effect of driver inhibitors alone by increasing the apoptotic response. CONCLUSIONS Inhibition of ATM reduced both glycolytic enzymes and OXPHOS levels in oncogene-driven cancer cells and enhanced apoptosis induced by driver inhibitors thus highlighting the possibility to use ATM and the driver inhibitors in combined regimens of anticancer therapy in vivo.
Collapse
Affiliation(s)
- Cristina Terlizzi
- Department of Advanced Biomedical Sciences, University "Federico II", 80131, Naples, Italy
| | - Viviana De Rosa
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Francesca Iommelli
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Antonio Pezone
- Department of Biology, University "Federico II", Naples, Italy
| | - Giovanna G Altobelli
- Department of Advanced Biomedical Sciences, University "Federico II", 80131, Naples, Italy
| | - Maurizio Maddalena
- Department of Advanced Biomedical Sciences, University "Federico II", 80131, Naples, Italy
| | - Jelena Dimitrov
- Department of Advanced Biomedical Sciences, University "Federico II", 80131, Naples, Italy
| | - Caterina De Rosa
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | | | - Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University "Federico II", 80131, Naples, Italy.
| |
Collapse
|
112
|
Shu K, Cai C, Chen W, Ding J, Guo Z, Wei Y, Zhang W. Prognostic value and immune landscapes of immunogenic cell death-associated lncRNAs in lung adenocarcinoma. Sci Rep 2023; 13:19151. [PMID: 37932413 PMCID: PMC10628222 DOI: 10.1038/s41598-023-46669-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023] Open
Abstract
Immunogenic cell death (ICD) has been demonstrated to activate T cells to kill tumor cells, which is closely related to tumor development, and long noncoding RNAs (lncRNAs) are also involved. However, it is not known whether ICD-related lncRNAs are associated with the development of lung adenocarcinoma (LUAD). We downloaded ICD-related genes from GeneCards and the transcriptome statistics of LUAD patients from The Cancer Genome Atlas (TCGA) and subsequently developed and verified a predictive model. A successful model was used together with other clinical features to construct a nomogram for predicting patient survival. To further study the mechanism of tumor action and to guide therapy, we performed enrichment analysis, tumor microenvironment analysis, somatic mutation analysis, drug sensitivity analysis and real-time quantitative polymerase chain reaction (RT-qPCR) analysis. Nine ICD-related lncRNAs with significant prognostic relevance were selected for model construction. Survival analysis demonstrated that overall survival was substantially shorter in the high-risk group than in the low-risk group (P < 0.001). This model was predictive of prognosis across all clinical subgroups. Cox regression analysis further supported the independent prediction ability of the model. Ultimately, a nomogram depending on stage and risk score was created and showed a better predictive performance than the nomogram without the risk score. Through enrichment analysis, the enriched pathways in the high-risk group were found to be primarily associated with metabolism and DNA replication. Tumor microenvironment analysis suggested that the immune cell concentration was lower in the high-risk group. Somatic mutation analysis revealed that the high-risk group contained more tumor mutations (P = 0.00018). Tumor immune dysfunction and exclusion scores exhibited greater sensitivity to immunotherapy in the high-risk group (P < 0.001). Drug sensitivity analysis suggested that the predictive model can also be applied to the choice of chemotherapy drugs. RT-qPCR analysis also validated the accuracy of the constructed model based on nine ICD-related lncRNAs. The prognostic model constructed based on the nine ICD-related lncRNAs showed good application value in assessing prognosis and guiding clinical therapy.
Collapse
Affiliation(s)
- Kexin Shu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chenxi Cai
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Wanying Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Jiatong Ding
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Zishun Guo
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Yiping Wei
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China.
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China.
| |
Collapse
|
113
|
Liu T, Ren Y, Wang Q, Wang Y, Li Z, Sun W, Fan D, Luan Y, Gao Y, Yan Z. Exploring the role of the disulfidptosis-related gene SLC7A11 in adrenocortical carcinoma: implications for prognosis, immune infiltration, and therapeutic strategies. Cancer Cell Int 2023; 23:259. [PMID: 37919768 PMCID: PMC10623781 DOI: 10.1186/s12935-023-03091-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Disulfidptosis and the disulfidptosis-related gene SLC7A11 have recently attracted significant attention for their role in tumorigenesis and tumour management. However, its association with adrenocortical carcinoma (ACC) is rarely discussed. METHODS Differential analysis, Cox regression analysis, and survival analysis were used to screen for the hub gene SLC7A11 in the TCGA and GTEx databases and disulfidptosis-related gene sets. Then, we performed an association analysis between SLC7A11 and clinically relevant factors in ACC patients. Univariate and multivariate Cox regression analyses were performed to evaluate the prognostic value of SLC7A11 and clinically relevant factors. Weighted gene coexpression analysis was used to find genes associated with SLC7A11. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses and the LinkedOmics database were used to analyse the functions of SLC7A11-associated genes. The CIBERSORT and Xcell algorithms were used to analyse the relationship between SLC7A11 and immune cell infiltration in ACC. The TISIDB database was applied to search for the correlation between SLC7A11 expression and immune chemokines. In addition, we performed a correlation analysis for SLC7A11 expression and tumour mutational burden and immune checkpoint-related genes and assessed drug sensitivity based on SLC7A11 expression. Immunohistochemistry and RT‒qPCR were used to validate the upregulation of SLC7A11 in the ACC. RESULTS SLC7A11 is highly expressed in multiple urological tumours, including ACC. SLC7A11 expression is strongly associated with clinically relevant factors (M-stage and MYL6 expression) in ACC. SLC7A11 and the constructed nomogram can accurately predict ACC patient outcomes. The functions of SLC7A11 and its closely related genes are tightly associated with the occurrence of disulfidptosis in ACC. SLC7A11 expression was tightly associated with various immune cell infiltration disorders in the ACC tumour microenvironment (TME). It was positively correlated with the expression of immune chemokines (CXCL8, CXCL3, and CCL20) and negatively correlated with the expression of immune chemokines (CXCL17 and CCL14). SLC7A11 expression was positively associated with the expression of immune checkpoint genes (NRP1, TNFSF4, TNFRSF9, and CD276) and tumour mutation burden. The expression level of SLC7A11 in ACC patients is closely associated withcthe drug sensitivity. CONCLUSION In ACC, high expression of SLC7A11 is associated with migration, invasion, drug sensitivity, immune infiltration disorders, and poor prognosis, and its induction of disulfidptosis is a promising target for the treatment of ACC.
Collapse
Affiliation(s)
- Tonghu Liu
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Yilin Ren
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Qixin Wang
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China
- Department of Surgery, Nanyang Central Hospital, 473005, Nanyang, Henan, China
| | - Yu Wang
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China
- Henan Engineering Research Center of Tumour Molecular Diagnosis and Treatment, 450001, Zhengzhou, Henan, China
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Zhiyuan Li
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Weibo Sun
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China
- Department of Radiation Oncology and Oncology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Dandan Fan
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China
- Henan Engineering Research Center of Tumour Molecular Diagnosis and Treatment, 450001, Zhengzhou, Henan, China
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Yongkun Luan
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China.
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China.
- Henan Engineering Research Center of Tumour Molecular Diagnosis and Treatment, 450001, Zhengzhou, Henan, China.
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China.
| | - Yukui Gao
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China.
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, 241001, Wuhu, Anhui, China.
| | - Zechen Yan
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China.
- Henan Engineering Research Center of Tumour Molecular Diagnosis and Treatment, 450001, Zhengzhou, Henan, China.
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China.
| |
Collapse
|
114
|
Yang F, Hilakivi-Clarke L, Shaha A, Wang Y, Wang X, Deng Y, Lai J, Kang N. Metabolic reprogramming and its clinical implication for liver cancer. Hepatology 2023; 78:1602-1624. [PMID: 36626639 PMCID: PMC10315435 DOI: 10.1097/hep.0000000000000005] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023]
Abstract
Cancer cells often encounter hypoxic and hypo-nutrient conditions, which force them to make adaptive changes to meet their high demands for energy and various biomaterials for biomass synthesis. As a result, enhanced catabolism (breakdown of macromolecules for energy production) and anabolism (macromolecule synthesis from bio-precursors) are induced in cancer. This phenomenon is called "metabolic reprogramming," a cancer hallmark contributing to cancer development, metastasis, and drug resistance. HCC and cholangiocarcinoma (CCA) are 2 different liver cancers with high intertumoral heterogeneity in terms of etiologies, mutational landscapes, transcriptomes, and histological representations. In agreement, metabolism in HCC or CCA is remarkably heterogeneous, although changes in the glycolytic pathways and an increase in the generation of lactate (the Warburg effect) have been frequently detected in those tumors. For example, HCC tumors with activated β-catenin are addicted to fatty acid catabolism, whereas HCC tumors derived from fatty liver avoid using fatty acids. In this review, we describe common metabolic alterations in HCC and CCA as well as metabolic features unique for their subsets. We discuss metabolism of NAFLD as well, because NAFLD will likely become a leading etiology of liver cancer in the coming years due to the obesity epidemic in the Western world. Furthermore, we outline the clinical implication of liver cancer metabolism and highlight the computation and systems biology approaches, such as genome-wide metabolic models, as a valuable tool allowing us to identify therapeutic targets and develop personalized treatments for liver cancer patients.
Collapse
Affiliation(s)
- Flora Yang
- BA/MD Joint Admission Scholars Program, University of Minnesota, Minneapolis, Minnesota
| | - Leena Hilakivi-Clarke
- Food Science and Nutrition Section, The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Aurpita Shaha
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Yuanguo Wang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Xianghu Wang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Yibin Deng
- Department of Urology, Masonic Cancer Center, The University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jinping Lai
- Department of Pathology and Laboratory Medicine, Kaiser Permanente Sacramento Medical Center, Sacramento, California
| | - Ningling Kang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| |
Collapse
|
115
|
Yuan M, Zhang X, Yue F, Zhang F, Jiang S, Zhou X, Lv J, Zhang Z, Sun Y, Chen Z, Wu H, Liu X, Yu X, Wei B, Jiang K, Lin F, Zuo Y, Ren S. CircNOLC1 Promotes Colorectal Cancer Liver Metastasis by Interacting with AZGP1 and Sponging miR-212-5p to Regulate Reprogramming of the Oxidative Pentose Phosphate Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205229. [PMID: 37870214 PMCID: PMC10667818 DOI: 10.1002/advs.202205229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 08/27/2023] [Indexed: 10/24/2023]
Abstract
Liver metastasis is a common cause of death in progressive colorectal cancer patients, but the molecular mechanisms remain unclear. Here, it is reported that a conserved and oxidative pentose phosphate pathway-associated circular RNA, circNOLC1, plays a crucial role in colorectal cancer liver metastasis. It is found that circNOLC1 silencing reduces the oxidative pentose phosphate pathway-related intermediate metabolites and elevates NADP+ /NADPH ratio and intracellular ROS levels, thereby attenuating colorectal cancer cell proliferation, migration, and liver metastasis. circNOLC1 interacting with AZGP1 to activate mTOR/SREBP1 signaling, or sponging miR-212-5p to upregulate c-Met expression, both of which can further induce G6PD to activate oxidative pentose phosphate pathway in colorectal cancer liver metastasis. Moreover, circNOLC1 is regulated by the transcription factor YY1 and specifically stabilized HuR induces its parental gene mRNA expression. The associations between circNOLC1 and these signaling molecules are validated in primary CRC and corresponding liver metastasis tissues. These findings reveal that circNOLC1 interacting with AZGP1 and circNOLC1/miR-212-5p/c-Met axis plays a key role in oxidative pentose phosphate pathway-mediated colorectal cancer liver metastasis, which may provide a novel target for precision medicine of colorectal cancer.
Collapse
Affiliation(s)
- Menglang Yuan
- Department of General SurgeryThe Second Hospital of Dalian Medical University116023DalianChina
- Department of OncologySidney Kimmel Comprehensive Cancer CenterSchool of MedicineJohns Hopkins UniversityBaltimoreMD21287USA
| | - Xinsheng Zhang
- Department of General SurgeryThe Second Hospital of Dalian Medical University116023DalianChina
| | - Fangxia Yue
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Feifan Zhang
- Department of General SurgeryThe Second Hospital of Dalian Medical University116023DalianChina
| | - Sufen Jiang
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Xu Zhou
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Jinjuan Lv
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Zhenyu Zhang
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Yuzhu Sun
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Zihao Chen
- Department of General SurgeryThe Second Hospital of Dalian Medical University116023DalianChina
| | - Han Wu
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Xiaoqian Liu
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Xiaoqi Yu
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Bowen Wei
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Kexin Jiang
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Fang Lin
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Yunfei Zuo
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Shuangyi Ren
- Department of General SurgeryThe Second Hospital of Dalian Medical University116023DalianChina
| |
Collapse
|
116
|
Wang H, Zhang J. The glucose metabolic reprogramming in hepatitis B virus infection and hepatitis B virus associated diseases. J Gastroenterol Hepatol 2023; 38:1886-1891. [PMID: 37654246 DOI: 10.1111/jgh.16340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 08/01/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
Hepatitis B virus (HBV) infection is closely related to viral hepatitis, liver cirrhosis, and hepatocellular carcinoma. HBV infection can reprogram metabolism processes of the host cells including glucose metabolism. The aberrant glucose metabolism may aid in viral infection and immune escape and may contribute to liver associated pathology. In this review, we discussed the interplay between HBV infection and glucose metabolism, which may provide new insights into HBV infection and pathology, novel intervention targets for HBV-related diseases.
Collapse
Affiliation(s)
- Hangle Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Ministry of Health (Peking University), Peking University Health Science Center, Beijing, China
| | - Jun Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Ministry of Health (Peking University), Peking University Health Science Center, Beijing, China
| |
Collapse
|
117
|
Davalieva K, Kiprijanovska S, Ivanovski O, Trifunovski A, Saidi S, Dimovski A, Popov Z. Proteomics Profiling of Bladder Cancer Tissues from Early to Advanced Stages Reveals NNMT and GALK1 as Biomarkers for Early Detection and Prognosis of BCa. Int J Mol Sci 2023; 24:14938. [PMID: 37834386 PMCID: PMC10573217 DOI: 10.3390/ijms241914938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 10/15/2023] Open
Abstract
The high recurrence rate and invasive diagnostic and monitoring methods in bladder cancer (BCa) clinical management require the development of new non-invasive molecular tools for early detection, particularly for low-grade and low-stage BCa as well as for risk stratification. By using an in-solution digestion method and label-free data-independent LC-MS/MS coupled with ion mobility, we profiled the BCa tissues from initiation to advanced stages and confidently identified and quantified 1619 proteins (≥2 peptides). A statistically significant difference in abundance (Anova ≤ 0.05) showed 494 proteins. Significant correlation with stage with steady up or down with BCa stages showed 15 proteins. Testing of NNMT, GALK1, and HTRA1 in urine samples showed excellent diagnostic potential for NNMT and GALK1 with AUC of 1.000 (95% CI: 1.000-1.000; p < 0.0001) and 0.801 (95% CI: 0.655-0.947; p < 0.0001), respectively. NNMT and GALK1 also showed very good potential in discriminating non-invasive low-grade from invasive high-grade BCa with AUC of 0.763 (95% CI: 0.606-0.921; p = 0.001) and 0.801 (95% CI: 0.653-0.950; p < 0.0001), respectively. The combination of NNMT and GALK1 increased prognostic accuracy (AUC = 0.813). Our results broaden the range of potential novel candidates for non-invasive BCa diagnosis and prognosis.
Collapse
Affiliation(s)
- Katarina Davalieva
- Research Centre for Genetic Engineering and Biotechnology “Georgi D Efremov”, Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia; (S.K.); (A.D.)
| | - Sanja Kiprijanovska
- Research Centre for Genetic Engineering and Biotechnology “Georgi D Efremov”, Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia; (S.K.); (A.D.)
| | - Ognen Ivanovski
- Clinical Centre “Mother Theresa”, University Clinic for Urology, 1000 Skopje, North Macedonia; (O.I.); (A.T.); (S.S.)
| | - Aleksandar Trifunovski
- Clinical Centre “Mother Theresa”, University Clinic for Urology, 1000 Skopje, North Macedonia; (O.I.); (A.T.); (S.S.)
| | - Skender Saidi
- Clinical Centre “Mother Theresa”, University Clinic for Urology, 1000 Skopje, North Macedonia; (O.I.); (A.T.); (S.S.)
| | - Aleksandar Dimovski
- Research Centre for Genetic Engineering and Biotechnology “Georgi D Efremov”, Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia; (S.K.); (A.D.)
- Faculty of Pharmacy, University “St. Cyril and Methodius”, 1000 Skopje, North Macedonia
| | - Zivko Popov
- Clinical Hospital “Acibadem Sistina”, 1000 Skopje, North Macedonia;
- Medical Faculty, University “St. Cyril and Methodius”, 1000 Skopje, North Macedonia
- Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia
| |
Collapse
|
118
|
Liu YK, Wu X, Hadisurya M, Li L, Kaimakliotis H, Iliuk A, Tao WA. One-Pot Analytical Pipeline for Efficient and Sensitive Proteomic Analysis of Extracellular Vesicles. J Proteome Res 2023; 22:3301-3310. [PMID: 37702715 PMCID: PMC10897859 DOI: 10.1021/acs.jproteome.3c00361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Extracellular vesicle (EV) proteomics emerges as an effective tool for discovering potential biomarkers for disease diagnosis, monitoring, and therapeutics. However, the current workflow of mass spectrometry-based EV proteome analysis is not fully compatible in a clinical setting due to inefficient EV isolation methods and a tedious sample preparation process. To streamline and improve the efficiency of EV proteome analysis, here we introduce a one-pot analytical pipeline integrating a robust EV isolation approach, EV total recovery and purification (EVtrap), with in situ protein sample preparation, to detect urinary EV proteome. By incorporating solvent-driven protein capture and fast on-bead digestion, the one-pot pipeline enabled the whole EV proteome analysis to be completed within one day. In comparison with the existing workflow, the one-pot pipeline was able to obtain better peptide yield and identify the equivalent number of unique EV proteins from 1 mL of urine. Finally, we applied the one-pot pipeline to profile proteomes in urinary EVs of bladder cancer patients. A total of 2774 unique proteins were identified in 53 urine samples using a 15 min gradient library-free data-independent acquisition method. Taken altogether, our novel one-pot analytical pipeline demonstrated its potential for routine and robust EV proteomics in biomedical applications.
Collapse
Affiliation(s)
- Yi-Kai Liu
- Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Xiaofeng Wu
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Marco Hadisurya
- Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Li Li
- Tymora Analytical Operations, West Lafayette, Indiana 47906, United States
| | - Hristos Kaimakliotis
- Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Anton Iliuk
- Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Tymora Analytical Operations, West Lafayette, Indiana 47906, United States
| | - W Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Tymora Analytical Operations, West Lafayette, Indiana 47906, United States
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
119
|
Pathak T, Benson JC, Johnson MT, Xin P, Abdelnaby AE, Walter V, Koltun WA, Yochum GS, Hempel N, Trebak M. Loss of STIM2 in colorectal cancer drives growth and metastasis through metabolic reprogramming and PERK-ATF4 endoplasmic reticulum stress pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560521. [PMID: 37873177 PMCID: PMC10592933 DOI: 10.1101/2023.10.02.560521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The endoplasmic reticulum (ER) stores large amounts of calcium (Ca2+), and the controlled release of ER Ca2+ regulates a myriad of cellular functions. Although altered ER Ca2+ homeostasis is known to induce ER stress, the mechanisms by which ER Ca2+ imbalance activate ER stress pathways are poorly understood. Stromal-interacting molecules STIM1 and STIM2 are two structurally homologous ER-resident Ca2+ sensors that synergistically regulate Ca2+ influx into the cytosol through Orai Ca2+ channels for subsequent signaling to transcription and ER Ca2+ refilling. Here, we demonstrate that reduced STIM2, but not STIM1, in colorectal cancer (CRC) is associated with poor patient prognosis. Loss of STIM2 causes SERCA2-dependent increase in ER Ca2+, increased protein translation and transcriptional and metabolic rewiring supporting increased tumor size, invasion, and metastasis. Mechanistically, STIM2 loss activates cMyc and the PERK/ATF4 branch of ER stress in an Orai-independent manner. Therefore, STIM2 and PERK/ATF4 could be exploited for prognosis or in targeted therapies to inhibit CRC tumor growth and metastasis.
Collapse
Affiliation(s)
- Trayambak Pathak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - J. Cory Benson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- UPMC Hillman Cancer Center. University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Martin T. Johnson
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Ping Xin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Ahmed Emam Abdelnaby
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Vonn Walter
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States
- Penn State Cancer Institute. The Pennsylvania State University College of Medicine, Hershey, United States
| | - Walter A. Koltun
- Department of Surgery, Division of Colon and Rectal Surgery, The Pennsylvania State University College of Medicine, Hershey, United States
| | - Gregory S. Yochum
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, United States
- Department of Surgery, Division of Colon and Rectal Surgery, The Pennsylvania State University College of Medicine, Hershey, United States
| | - Nadine Hempel
- UPMC Hillman Cancer Center. University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- UPMC Hillman Cancer Center. University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
120
|
Hirose Y, Taniguchi K. Intratumoral metabolic heterogeneity of colorectal cancer. Am J Physiol Cell Physiol 2023; 325:C1073-C1084. [PMID: 37661922 DOI: 10.1152/ajpcell.00139.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/31/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
Although the metabolic phenotype within tumors is known to differ significantly from that of the surrounding normal tissue, the importance of this heterogeneity is just becoming widely recognized. Colorectal cancer (CRC) is often classified as the Warburg phenotype, a metabolic type in which the glycolytic system is predominant over oxidative phosphorylation (OXPHOS) in mitochondria for energy production. However, this dichotomy (glycolysis vs. OXPHOS) may be too simplistic and not accurately represent the metabolic characteristics of CRC. Therefore, in this review, we decompose metabolic phenomena into factors based on their source/origin and reclassify them into two categories: extrinsic and intrinsic. In the CRC context, extrinsic factors include those based on the environment, such as hypoxia, nutrient deprivation, and the tumor microenvironment, whereas intrinsic factors include those based on subpopulations, such as pathological subtypes and cancer stem cells. These factors form multiple layers inside and outside the tumor, affecting them additively, dominantly, or mutually exclusively. Consequently, the metabolic phenotype is a heterogeneous and fluid phenomenon reflecting the spatial distribution and temporal continuity of these factors. This allowed us to redefine the characteristics of specific metabolism-related factors in CRC and summarize and update our accumulated knowledge of their heterogeneity. Furthermore, we positioned tumor budding in CRC as an intrinsic factor and a novel form of metabolic heterogeneity, and predicted its metabolic dynamics, noting its similarity to circulating tumor cells and epithelial-mesenchymal transition. Finally, the possibilities and limitations of using human tumor tissue as research material to investigate and assess metabolic heterogeneity are discussed.
Collapse
Affiliation(s)
- Yoshinobu Hirose
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Kohei Taniguchi
- Division of Translational Research, Center for Medical Research & Development, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| |
Collapse
|
121
|
Delorme-Axford E, Wen X, Klionsky DJ. The yeast transcription factor Stb5 acts as a negative regulator of autophagy by modulating cellular metabolism. Autophagy 2023; 19:2719-2732. [PMID: 37345792 PMCID: PMC10472870 DOI: 10.1080/15548627.2023.2228533] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 06/02/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023] Open
Abstract
Macroautophagy/autophagy is a highly conserved pathway of cellular degradation and recycling that maintains cell health during homeostatic conditions and facilitates survival during stress. Aberrant cellular autophagy contributes to the pathogenesis of human diseases such as cancer, neurodegeneration, and cardiovascular, metabolic and lysosomal storage disorders. Despite decades of research, there remain unanswered questions as to how autophagy modulates cellular metabolism, and, conversely, how cellular metabolism affects autophagy activity. Here, we have identified the yeast metabolic transcription factor Stb5 as a negative regulator of autophagy. Chromosomal deletion of STB5 in the yeast Saccharomyces cerevisiae enhances autophagy. Loss of Stb5 results in the upregulation of select autophagy-related (ATG) transcripts under nutrient-replete conditions; however, the Stb5-mediated impact on autophagy occurs primarily through its effect on genes involved in NADPH production and the pentose phosphate pathway. This work provides insight into the intersection of Stb5 as a transcription factor that regulates both cellular metabolic responses and autophagy activity.Abbreviations: bp, base pairs; ChIP, chromatin immunoprecipitation; G6PD, glucose-6-phosphate dehydrogenase; GFP, green fluorescent protein; IDR, intrinsically disordered region; NAD, nicotinamide adenine dinucleotide; NADP+, nicotinamide adenine dinucleotide phosphate; NADPH, nicotinamide adenine dinucleotide phosphate (reduced); ORF, open reading frame; PA, protein A; PCR, polymerase chain reaction; PE, phosphatidylethanolamine; PPP, pentose phosphate pathway; prApe1, precursor aminopeptidase I; ROS, reactive oxygen species; RT-qPCR, real-time quantitative PCR; SD, standard deviation; TF, transcription factor; TOR, target of rapamycin; WT, wild-type.
Collapse
Affiliation(s)
| | - Xin Wen
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel J. Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
122
|
Wang Z, Guo X, Lian J, Ji Y, Li K. Prognostic value of amino acid metabolism-related gene expression in invasive breast carcinoma. J Cancer Res Clin Oncol 2023; 149:11117-11133. [PMID: 37340191 DOI: 10.1007/s00432-023-04985-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/13/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND An increasing number of studies indicated that metabolic reprogramming of amino acid metabolism may either promote or inhibit tumor progression. The purpose of this study was to investigate the ability of a gene risk signature associated with amino acid metabolism to predict the prognosis and immune characteristics of invasive breast carcinoma. METHODS LASSO Cox regression analysis was performed to construct and validate the prognostic risk signature based on the expression of 9 amino acid metabolism-related genes. The predictive value of the signature, immune characteristics, and chemotherapeutic drugs was also predicted. Finally, 9 significant genes were examined in MDA-MB-231 and MCF-7 cells, and the predicted chemotherapeutic drugs were also verified. RESULTS The prognosis of the low-risk group was better than that of the high-risk group. The areas under the curve (AUCs) at 1, 2, and 3 years were 0.852, 0.790, and 0.736, respectively. In addition, the GSEA results for KEGG and GO revealed that samples with a high-risk score exhibited a variety of highly malignant manifestations. The high-risk group was characterized by an increased number of M2 macrophages, a high level of tumor purity, low levels of APC co-stimulation, cytolytic activity, HLA, para-inflammation, and type I IFN response. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) confirmed that MDA-MB-231 and MCF-7 cells express 9 amino acid metabolism-related genes differently. In addition, cell experiments were conducted to examine the effect of cephaeline-induced on cell viability, migration ability, and protein expression of the PI3K/AKT signaling pathway and HIF-1α. CONCLUSION We established a risk signature based on 9 amino acid metabolism-related genes for invasive breast carcinoma. Further analyses revealed that this risk signature is superior to other clinical indexes in survival prediction and that the subgroups identified by the risk signature exhibit distinct immune characteristics. Cephaeline was determined to be a superior option for patients in high-risk groups.
Collapse
Affiliation(s)
- Zilin Wang
- Department of Radiology, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, No. 650 New Songjiang Road, Shanghai, 200080, People's Republic of China
| | - Xinyu Guo
- Department of Radiology, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, No. 650 New Songjiang Road, Shanghai, 200080, People's Republic of China
| | - Jingge Lian
- Department of Radiology, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, No. 650 New Songjiang Road, Shanghai, 200080, People's Republic of China
| | - Ying Ji
- Department of Radiology, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, No. 650 New Songjiang Road, Shanghai, 200080, People's Republic of China
| | - Kangan Li
- Department of Radiology, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, No. 650 New Songjiang Road, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
123
|
Zyla J, Papiez A, Zhao J, Qu R, Li X, Kluger Y, Polanska J, Hatzis C, Pusztai L, Marczyk M. Evaluation of zero counts to better understand the discrepancies between bulk and single-cell RNA-Seq platforms. Comput Struct Biotechnol J 2023; 21:4663-4674. [PMID: 37841335 PMCID: PMC10568495 DOI: 10.1016/j.csbj.2023.09.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023] Open
Abstract
Recent advances in sample preparation and sequencing technology have made it possible to profile the transcriptomes of individual cells using single-cell RNA sequencing (scRNA-Seq). Compared to bulk RNA-Seq data, single-cell data often contain a higher percentage of zero reads, mainly due to lower sequencing depth per cell, which affects mostly measurements of low-expression genes. However, discrepancies between platforms are observed regardless of expression level. Using four paired datasets with multiple samples each, we investigated technical and biological factors that can contribute to this expression shift. Using two separate machine learning models we found that, in addition to expression level, RNA integrity, gene or UTR3 length, and the number of transcripts potentially also influence the occurrence of zeros. These findings could enable the development of novel analytical methods for cross-platform expression shift correction. We also identified genes and biological pathways in our diverse datasets that consistently showed differences when assessed at the single cell versus bulk level to assist in interpreting analysis across transcriptomic platforms. At the gene level, 25 genes (0.12%) were found in all datasets as discordant, but at the pathway level, 7 pathways (2.02%) showed shared enrichment in discordant genes.
Collapse
Affiliation(s)
- Joanna Zyla
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice 44-100, Poland
| | - Anna Papiez
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice 44-100, Poland
| | - Jun Zhao
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT 06510, USA
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Rihao Qu
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT 06510, USA
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Xiaotong Li
- Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yuval Kluger
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT 06510, USA
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
- Applied Mathematics Program, Yale University, New Haven, CT, USA
| | - Joanna Polanska
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice 44-100, Poland
| | - Christos Hatzis
- Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Lajos Pusztai
- Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Michal Marczyk
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice 44-100, Poland
- Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
124
|
Zhang W, Zhou H, Li H, Mou H, Yinwang E, Xue Y, Wang S, Zhang Y, Wang Z, Chen T, Sun H, Wang F, Zhang J, Chai X, Chen S, Li B, Zhang C, Gao J, Ye Z. Cancer cells reprogram to metastatic state through the acquisition of platelet mitochondria. Cell Rep 2023; 42:113147. [PMID: 37756158 DOI: 10.1016/j.celrep.2023.113147] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/30/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Metastasis is the major cause of cancer deaths, and cancer cells evolve to adapt to various tumor microenvironments, which hinders the treatment of tumor metastasis. Platelets play critical roles in tumor development, especially during metastasis. Here, we elucidate the role of platelet mitochondria in tumor metastasis. Cancer cells are reprogrammed to a metastatic state through the acquisition of platelet mitochondria via the PINK1/Parkin-Mfn2 pathway. Furthermore, platelet mitochondria regulate the GSH/GSSG ratio and reactive oxygen species (ROS) in cancer cells to promote lung metastasis of osteosarcoma. Impairing platelet mitochondrial function has proven to be an efficient approach to impair metastasis, providing a direction for osteosarcoma therapy. Our findings demonstrate mitochondrial transfer between platelets and cancer cells and suggest a role for platelet mitochondria in tumor metastasis.
Collapse
Affiliation(s)
- Wenkan Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hao Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hengyuan Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Haochen Mou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Eloy Yinwang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yucheng Xue
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shengdong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yongxing Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zenan Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tao Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hangxiang Sun
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Fangqian Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jiahao Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xupeng Chai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shixin Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Binghao Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China; Institute of Orthopedic Research, Zhejiang University, Hangzhou 310009, People's Republic of China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
125
|
Zhang Q, Song Q, Liu S, Xu Y, Gao D, Lu P, Liu Y, Zhao G, Wu L, Zhao C, Yang J. Integrated transcriptomic and metabolomic analysis reveals the metabolic programming of GM-CSF- and M-CSF- differentiated mouse macrophages. Front Immunol 2023; 14:1230772. [PMID: 37818352 PMCID: PMC10560851 DOI: 10.3389/fimmu.2023.1230772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Macrophages play a critical role in the inflammatory response and tumor development. Macrophages are primarily divided into pro-inflammatory M1-like and anti-inflammatory M2-like macrophages based on their activation status and functions. In vitro macrophage models could be derived from mouse bone marrow cells stimulated with two types of differentiation factors: GM-CSF (GM-BMDMs) and M-CSF (M-BMDMs), to represent M1- and M2-like macrophages, respectively. Since macrophage differentiation requires coordinated metabolic reprogramming and transcriptional rewiring in order to fulfill their distinct roles, we combined both transcriptome and metabolome analysis, coupled with experimental validation, to gain insight into the metabolic status of GM- and M-BMDMs. The data revealed higher levels of the tricarboxylic acid cycle (TCA cycle), oxidative phosphorylation (OXPHOS), fatty acid oxidation (FAO), and urea and ornithine production from arginine in GM-BMDMs, and a preference for glycolysis, fatty acid storage, bile acid metabolism, and citrulline and nitric oxide (NO) production from arginine in M-BMDMs. Correlation analysis with the proteomic data showed high consistency in the mRNA and protein levels of metabolic genes. Similar results were also obtained when compared to RNA-seq data of human monocyte derived macrophages from the GEO database. Furthermore, canonical macrophage functions such as inflammatory response and phagocytosis were tightly associated with the representative metabolic pathways. In the current study, we identified the core metabolites, metabolic genes, and functional terms of the two distinct mouse macrophage populations. We also distinguished the metabolic influences of the differentiation factors GM-CSF and M-CSF, and wish to provide valuable information for in vitro macrophage studies.
Collapse
Affiliation(s)
- Qianyue Zhang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Qiaoling Song
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Innovation Platform of Marine Drug Screening and Evaluation, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Shan Liu
- Innovation Platform of Marine Drug Screening and Evaluation, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Yuting Xu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Danling Gao
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Peizhe Lu
- Department of Neuroscience, University of Michigan, Ann Arbor, MI, United States
| | - Yuantao Liu
- Department of Endocrinology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Guanghui Zhao
- Medical Laboratory Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Oncology Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Lihong Wu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Chenyang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Innovation Platform of Marine Drug Screening and Evaluation, Qingdao Marine Science and Technology Center, Qingdao, China
| |
Collapse
|
126
|
Sun N, Zhang Y, Dong J, Liu G, Liu Z, Wang J, Qiao Z, Zhang J, Duan K, Nian X, Ma Z, Yang X. Metabolomics profiling reveals differences in proliferation between tumorigenic and non-tumorigenic Madin-Darby canine kidney (MDCK) cells. PeerJ 2023; 11:e16077. [PMID: 37744241 PMCID: PMC10517658 DOI: 10.7717/peerj.16077] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 08/20/2023] [Indexed: 09/26/2023] Open
Abstract
Background Madin-Darby canine kidney (MDCK) cells are a cellular matrix in the production of influenza vaccines. The proliferation rate of MDCK cells is one of the critical factors that determine the vaccine production cycle. It is yet to be determined if there is a correlation between cell proliferation and alterations in metabolic levels. This study aimed to explore the metabolic differences between MDCK cells with varying proliferative capabilities through the use of both untargeted and targeted metabolomics. Methods To investigate the metabolic discrepancies between adherent cell groups (MDCK-M60 and MDCK-CL23) and suspension cell groups (MDCK-XF04 and MDCK-XF06), untargeted and targeted metabolomics were used. Utilizing RT-qPCR analysis, the mRNA expressions of key metabolites enzymes were identified. Results An untargeted metabolomics study demonstrated the presence of 81 metabolites between MDCK-M60 and MDCK-CL23 cells, which were mainly affected by six pathways. An analysis of MDCK-XF04 and MDCK-XF06 cells revealed a total of 113 potential metabolites, the majority of which were impacted by ten pathways. Targeted metabolomics revealed a decrease in the levels of choline, tryptophan, and tyrosine in MDCK-CL23 cells, which was in accordance with the results of untargeted metabolomics. Additionally, MDCK-XF06 cells experienced a decrease in 5'-methylthioadenosine and tryptophan, while S-adenosylhomocysteine, kynurenine, 11Z-eicosenoic acid, 3-phosphoglycerate, glucose 6-phosphate, and phosphoenolpyruvic acid concentrations were increased. The mRNA levels of MAT1A, MAT2B, IDO1, and IDO2 in the two cell groups were all increased, suggesting that S-adenosylmethionine and tryptophan may have a significant role in cell metabolism. Conclusions This research examines the effect of metabolite fluctuations on cell proliferation, thus offering a potential way to improve the rate of MDCK cell growth.
Collapse
Affiliation(s)
- Na Sun
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Lanzhou, China
| | - Yuchuan Zhang
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Jian Dong
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Geng Liu
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Zhenbin Liu
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Lanzhou, China
| | - Jiamin Wang
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Lanzhou, China
- Gansu Provincial Bioengineering Materials Engineering Research Center, Lanzhou, China
| | - Zilin Qiao
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Lanzhou, China
- Gansu Provincial Bioengineering Materials Engineering Research Center, Lanzhou, China
| | - Jiayou Zhang
- Wuhan Institute of Biological Products Co., Ltd., Wuhan, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan, China
| | - Kai Duan
- Wuhan Institute of Biological Products Co., Ltd., Wuhan, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan, China
| | - Xuanxuan Nian
- Wuhan Institute of Biological Products Co., Ltd., Wuhan, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan, China
| | - Zhongren Ma
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Lanzhou, China
- Key Laboratory of Biotechnology and Bioengineering of National Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Xiaoming Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan, China
- China National Biotech Group Company Limited, Beijing, China
| |
Collapse
|
127
|
Corder ML, Petricoin EF, Li Y, Cleland TP, DeCandia AL, Alonso Aguirre A, Pukazhenthi BS. Metabolomic profiling implicates mitochondrial and immune dysfunction in disease syndromes of the critically endangered black rhinoceros (Diceros bicornis). Sci Rep 2023; 13:15464. [PMID: 37726331 PMCID: PMC10509206 DOI: 10.1038/s41598-023-41508-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023] Open
Abstract
The critically endangered black rhinoceros (Diceros bicornis; black rhino) experiences extinction threats from poaching in-situ. The ex-situ population, which serves as a genetic reservoir against impending extinction threats, experiences its own threats to survival related to several disease syndromes not typically observed among their wild counterparts. We performed an untargeted metabolomic analysis of serum from 30 ex-situ housed black rhinos (Eastern black rhino, EBR, n = 14 animals; Southern black rhino, SBR, n = 16 animals) and analyzed differences in metabolite profiles between subspecies, sex, and health status (healthy n = 13 vs. diseased n = 14). Of the 636 metabolites detected, several were differentially (fold change > 1.5; p < 0.05) expressed between EBR vs. SBR (40 metabolites), female vs. male (36 metabolites), and healthy vs. diseased (22 metabolites). Results suggest dysregulation of propanoate, amino acid metabolism, and bile acid biosynthesis in the subspecies and sex comparisons. Assessment of healthy versus diseased rhinos indicates involvement of arachidonic acid metabolism, bile acid biosynthesis, and the pentose phosphate pathway in animals exhibiting inflammatory disease syndromes. This study represents the first systematic characterization of the circulating serum metabolome in the black rhinoceros. Findings further implicate mitochondrial and immune dysfunction as key contributors for the diverse disease syndromes reported in ex-situ managed black rhinos.
Collapse
Affiliation(s)
- Molly L Corder
- Smithsonian's National Zoo and Conservation Biology Institute, Center for Species Survival, Front Royal, 22630, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, 20900, USA
- Department of Environmental Sciences and Policy, George Mason University, Fairfax, Virginia, 22030, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, 20900, USA
| | - Yue Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20742, USA
| | | | - Alexandra L DeCandia
- Department of Biology, Georgetown University, Washington, DC, 20057, USA
- Smithsonian's National Zoo and Conservation Biology Institute, Center for Conservation Genomics, Washington, DC, 20008, USA
| | - A Alonso Aguirre
- Department of Fish, Wildlife, and Conservation Biology, Warner College of Natural Resources, Colorado State University, Fort Collins, 80523, USA
| | - Budhan S Pukazhenthi
- Smithsonian's National Zoo and Conservation Biology Institute, Center for Species Survival, Front Royal, 22630, USA.
| |
Collapse
|
128
|
Aboushousha R, van der Velden J, Hamilton N, Peng Z, MacPherson M, Erickson C, White S, Wouters EFM, Reynaert NL, Seward DJ, Li J, Janssen-Heininger YMW. Glutaredoxin attenuates glutathione levels via deglutathionylation of Otub1 and subsequent destabilization of system x C. SCIENCE ADVANCES 2023; 9:eadi5192. [PMID: 37703360 PMCID: PMC10499329 DOI: 10.1126/sciadv.adi5192] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/10/2023] [Indexed: 09/15/2023]
Abstract
Glutathione (GSH) is a critical component of the cellular redox system that combats oxidative stress. The glutamate-cystine antiporter, system xC-, is a key player in GSH synthesis that allows for the uptake of cystine, the rate-limiting building block of GSH. It is unclear whether GSH or GSH-dependent protein oxidation [protein S-glutathionylation (PSSG)] regulates the activity of system xC-. We demonstrate that an environment of enhanced PSSG promotes GSH increases via a system xC--dependent mechanism. Absence of the deglutathionylase, glutaredoxin (GLRX), augmented SLC7A11 protein and led to significant increases of GSH content. S-glutathionylation of C23 or C204 of the deubiquitinase OTUB1 promoted interaction with the E2-conjugating enzyme UBCH5A, leading to diminished ubiquitination and proteasomal degradation of SLC7A11 and augmentation of GSH, effects that were reversed by GLRX. These findings demonstrate an intricate link between GLRX and GSH via S-glutathionylation of OTUB1 and system xC- and illuminate a previously unknown feed-forward regulatory mechanism whereby enhanced GSH protein oxidation augments cellular GSH.
Collapse
Affiliation(s)
- Reem Aboushousha
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jos van der Velden
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Nicholas Hamilton
- Department of Chemistry, University of Vermont, Burlington, VT 05405, USA
| | - Zhihua Peng
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Maximilian MacPherson
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Cuixia Erickson
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Sheryl White
- Department of Neurological Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Emiel F. M. Wouters
- Department of Respiratory Medicine, NUTRIM School of nutrition and translational research in metabolism, Maastricht University Medical Center, Maastricht, Netherlands
- Ludwig Boltzmann Institute for Lung Research, Vienna, Austria
| | - Niki L. Reynaert
- Department of Respiratory Medicine, NUTRIM School of nutrition and translational research in metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - David J. Seward
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jianing Li
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
129
|
Yan R, Lin B, Jin W, Tang L, Hu S, Cai R. NRF2, a Superstar of Ferroptosis. Antioxidants (Basel) 2023; 12:1739. [PMID: 37760042 PMCID: PMC10525540 DOI: 10.3390/antiox12091739] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Ferroptosis is an iron-dependent and lipid peroxidation-driven cell death cascade, occurring when there is an imbalance of redox homeostasis in the cell. Nuclear factor erythroid 2-related factor 2 (NFE2L2, also known as NRF2) is key for cellular antioxidant responses, which promotes downstream genes transcription by binding to their antioxidant response elements (AREs). Numerous studies suggest that NRF2 assumes an extremely important role in the regulation of ferroptosis, for its various functions in iron, lipid, and amino acid metabolism, and so on. Many pathological states are relevant to ferroptosis. Abnormal suppression of ferroptosis is found in many cases of cancer, promoting their progression and metastasis. While during tissue damages, ferroptosis is recurrently promoted, resulting in a large number of cell deaths and even dysfunctions of the corresponding organs. Therefore, targeting NRF2-related signaling pathways, to induce or inhibit ferroptosis, has become a great potential therapy for combating cancers, as well as preventing neurodegenerative and ischemic diseases. In this review, a brief overview of the research process of ferroptosis over the past decade will be presented. In particular, the mechanisms of ferroptosis and a focus on the regulation of ferroptosis by NRF2 will be discussed. Finally, the review will briefly list some clinical applications of targeting the NRF2 signaling pathway in the treatment of diseases.
Collapse
Affiliation(s)
| | | | | | | | - Shuming Hu
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (R.Y.); (B.L.); (W.J.); (L.T.)
| | - Rong Cai
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (R.Y.); (B.L.); (W.J.); (L.T.)
| |
Collapse
|
130
|
Akossi RF, Delbac F, El Alaoui H, Wawrzyniak I, Peyretaillade E. The intracellular parasite Anncaliia algerae induces a massive miRNA down-regulation in human cells. Noncoding RNA Res 2023; 8:363-375. [PMID: 37275245 PMCID: PMC10238475 DOI: 10.1016/j.ncrna.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/27/2023] [Accepted: 05/07/2023] [Indexed: 06/07/2023] Open
Abstract
Anncaliia algerae belongs to microsporidia, a group of obligate intracellular parasites related to fungi. These parasites are largely spread in water and food-webs and can infect a wide variety of hosts ranging from invertebrates to vertebrates including humans. In humans, microsporidian infections are mainly opportunistic as immunocompetent hosts can clear parasites naturally. Recent studies however have reported persistent microsporidian infections and have highlighted them as a risk factor in colon cancer. This may be a direct result of cell infection or may be an indirect effect of the infectious microenvironment and the host's response. In both cases, this raises the question of the effects of microsporidian infection at the host and host-cell levels. We aimed to address the question of human host intracellular response to microsporidian infection through a transcriptomic kinetic study of human foreskin fibroblasts (HFF) infected with A.algerae, a human infecting microsporidia with an exceptionally wide host range. We focused solely on host response studying both coding and small non-coding miRNA expression. Our study revealed a generalized down-regulation of cell miRNAs throughout infection with up to 547 different miRNAs downregulated at some timepoints and also transcriptomic dysregulations that could facilitate parasite development with immune and lipid metabolism genes modulation. We also hypothesize possible small nucleic acid expropriation explaining the miRNA downregulation. This work contributes to a better understanding of the dialogue that can occur between an intracellular parasite and its host at the cellular level, and can guide future studies on microsporidian infection biology to unravel the mode of action of these minimalist parasites at the tissue or host levels.We have also generated a kinetic and comprehensive transcriptomic data set of an infectious process that can help support comparative studies in the broader field of parasitology. Lastly, these results may warrant for caution regarding microsporidian exposure and persistent infections.
Collapse
Affiliation(s)
- Reginald Florian Akossi
- Laboratoire “Microorganismes: Génome et Environnement” (LMGE), UMR 6023, Université Clermont Auvergne and CNRS, F-63000, Clermont-Ferrand, France
| | - Fréderic Delbac
- Laboratoire “Microorganismes: Génome et Environnement” (LMGE), UMR 6023, Université Clermont Auvergne and CNRS, F-63000, Clermont-Ferrand, France
| | - Hicham El Alaoui
- Laboratoire “Microorganismes: Génome et Environnement” (LMGE), UMR 6023, Université Clermont Auvergne and CNRS, F-63000, Clermont-Ferrand, France
| | - Ivan Wawrzyniak
- Laboratoire “Microorganismes: Génome et Environnement” (LMGE), UMR 6023, Université Clermont Auvergne and CNRS, F-63000, Clermont-Ferrand, France
| | - Eric Peyretaillade
- Laboratoire “Microorganismes: Génome et Environnement” (LMGE), UMR 6023, Université Clermont Auvergne and CNRS, F-63000, Clermont-Ferrand, France
| |
Collapse
|
131
|
Hao Y, Ren T, Huang X, Li M, Lee JH, Chen Q, Liu R, Tang Q. Rapid phosphorylation of glucose-6-phosphate dehydrogenase by casein kinase 2 sustains redox homeostasis under ionizing radiation. Redox Biol 2023; 65:102810. [PMID: 37478541 PMCID: PMC10404535 DOI: 10.1016/j.redox.2023.102810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/24/2023] [Accepted: 07/06/2023] [Indexed: 07/23/2023] Open
Abstract
Exposure to ionizing radiation leads to oxidative damages in living cells. NADPH provides the indispensable reducing power to regenerate the reduced glutathione to maintain cellular redox equilibria. In mammalian cells, pentose phosphate pathway (PPP) is the major route to produce NADPH by using glycolytic intermediates, and the rate-limiting step of PPP is controlled by glucose-6-phosphate dehydrogenase (G6PD). Nevertheless, whether G6PD is timely co-opted under ionizing radiation to cope with oxidative stress remains elusive. Here we show that cellular G6PD activity is induced 30 min after ionizing radiation, while its protein expression is mostly unchanged. Mechanistically, casein kinase 2 (CK2) phosphorylates G6PD T145 under ionizing radiation, which consolidates the enzymatic activity of G6PD by facilitating G6PD binding with its substrate NADP+. Further, CK2-dependent G6PD T145 phosphorylation promotes NADPH production, decreases ROS level and supports cell proliferation under ionizing radiation. Our findings report a new anti-oxidative signaling route under ionizing radiation, by which CK2-mediated rapid activation of G6PD orchestrates NADPH synthesis to maintain redox homeostasis, thereby highlighting its potential value in the early treatment of ionizing radiation-induced injuries.
Collapse
Affiliation(s)
- Yilong Hao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, PR China
| | - Tao Ren
- Oncology Department (Key Clinical Specialty of Sichuan Province), The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, PR China
| | - Xiaoke Huang
- Department of Urology, Xindu District People's Hospital of Chengdu, Chengdu, 610500, China
| | - Mi Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jong-Ho Lee
- Department of Health Sciences, The Graduated School of Dong-A University, Busan, 49315, Republic of Korea
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, PR China.
| | - Rui Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, PR China.
| | - Qingfeng Tang
- Department of Urology, Xindu District People's Hospital of Chengdu, Chengdu, 610500, China.
| |
Collapse
|
132
|
Kawakami H, Chen KQ, Zhang R, Pappas MP, Bailey A, Reisz JA, Corcoran D, Nishinakamura R, D'Alessandro A, Kawakami Y. Sall4 restricts glycolytic metabolism in limb buds through transcriptional regulation of glycolytic enzyme genes. Dev Biol 2023; 501:28-38. [PMID: 37301463 PMCID: PMC10330914 DOI: 10.1016/j.ydbio.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/09/2023] [Accepted: 06/07/2023] [Indexed: 06/12/2023]
Abstract
Recent studies illustrate the importance of regulation of cellular metabolism, especially glycolysis and pathways branching from glycolysis, during vertebrate embryo development. For example, glycolysis generates cellular energy ATP. Glucose carbons are also directed to the pentose phosphate pathway, which is needed to sustain anabolic processes in the rapidly growing embryos. However, our understanding of the exact status of glycolytic metabolism as well as genes that regulate glycolytic metabolism are still incomplete. Sall4 is a zinc finger transcription factor that is highly expressed in undifferentiated cells in developing mouse embryos, such as blastocysts and the post-implantation epiblast. TCre; Sall4 conditional knockout mouse embryos exhibit various defects in the posterior part of the body, including hindlimbs. Using transcriptomics approaches, we found that many genes encoding glycolytic enzymes are upregulated in the posterior trunk, including the hindlimb-forming region, of Sall4 conditional knockout mouse embryos. In situ hybridization and qRT-PCR also confirmed upregulation of expression of several glycolytic genes in hindlimb buds. A fraction of those genes are bound by SALL4 at the promoters, gene bodies or distantly-located regions, suggesting that Sall4 directly regulates expression of several glycolytic enzyme genes in hindlimb buds. To further gain insight into the metabolic status associated with the observed changes at the transcriptional level, we performed a comprehensive analysis of metabolite levels in limb buds in wild type and Sall4 conditional knockout embryos by high-resolution mass spectrometry. We found that the levels of metabolic intermediates of glycolysis are lower, but glycolytic end-products pyruvate and lactate did not exhibit differences in Sall4 conditional knockout hindlimb buds. The increased expression of glycolytic genes would have caused accelerated glycolytic flow, resulting in low levels of intermediates. This condition may have prevented intermediates from being re-directed to other pathways, such as the pentose phosphate pathway. Indeed, the change in glycolytic metabolite levels is associated with reduced levels of ATP and metabolites of the pentose phosphate pathway. To further test whether glycolysis regulates limb patterning downstream of Sall4, we conditionally inactivated Hk2, which encodes a rate-limiting enzyme gene in glycolysis and is regulated by Sall4. The TCre; Hk2 conditional knockout hindlimb exhibited a short femur, and a lack of tibia and anterior digits in hindlimbs, which are defects similarly found in the TCre; Sall4 conditional knockout. The similarity of skeletal defects in Sall4 mutants and Hk2 mutants suggests that regulation of glycolysis plays a role in hindlimb patterning. These data suggest that Sall4 restricts glycolysis in limb buds and contributes to patterning and regulation of glucose carbon flow during development of limb buds.
Collapse
Affiliation(s)
- Hiroko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA; Developmental Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - Katherine Q Chen
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Ruizhi Zhang
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Matthew P Pappas
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Abigail Bailey
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dylan Corcoran
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA; Developmental Biology Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
133
|
Zhang L, Jiao G, You Y, Li X, Liu J, Sun Z, Li Q, Dai Z, Ma J, Zhou H, Li G, Meng C, Chen Y. Arginine methylation of PPP1CA by CARM1 regulates glucose metabolism and affects osteogenic differentiation and osteoclastic differentiation. Clin Transl Med 2023; 13:e1369. [PMID: 37649137 PMCID: PMC10468565 DOI: 10.1002/ctm2.1369] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND The imbalance between osteoblasts and osteoclasts may lead to osteoporosis. Osteoblasts and osteoclasts have different energy requirements, with aerobic glycolysis being the prominent metabolic feature of osteoblasts, while osteoclast differentiation and fusion are driven by oxidative phosphorylation. METHODS By polymerase chain reaction as well as Western blotting, we assayed coactivator-associated arginine methyltransferase 1 (CARM1) expression in bone tissue, the mouse precranial osteoblast cell line MC3T3-E1 and the mouse monocyte macrophage leukaemia cell line RAW264.7, and expression of related genes during osteogenic differentiation and osteoclast differentiation. Using gene overexpression (lentivirus) and loss-of-function approach (CRISPR/Cas9-mediated knockout) in vitro, we examined whether CARM1 regulates osteogenic differentiation and osteoblast differentiation by metabolic regulation. Transcriptomic assays and metabolomic assays were used to find the mechanism of action of CARM1. Furthermore, in vitro methylation assays were applied to clarify the arginine methylation site of PPP1CA by CARM1. RESULTS We discovered that CARM1 reprogrammed glucose metabolism in osteoblasts and osteoclasts from oxidative phosphorylation to aerobic glycolysis, thereby promoting osteogenic differentiation and inhibiting osteoclastic differentiation. In vivo experiments revealed that CARM1 significantly decreased bone loss in osteoporosis model mice. Mechanistically, CARM1 methylated R23 of PPP1CA, affected the dephosphorylation of AKT-T450 and AMPK-T172, and increased the activities of phosphofructokinase-1 and pructose-2,6-biphosphatase3, causing an up-regulation of glycolytic flux. At the same time, as a transcriptional coactivator, CARM1 regulated the expression of pyruvate dehydrogenase kinase 3, which resulted in the inhibition of pyruvate dehydrogenase activity and inhibition of the tricarboxylic acid cycle, leading to a subsequent decrease in the flux of oxidative phosphorylation. CONCLUSIONS These findings reveal for the first time the mechanism by which CARM1 affects both osteogenesis and osteoclast differentiation through metabolic regulation, which may represent a new feasible treatment strategy for osteoporosis.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of MicroorthopaedicsAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanShandongChina
- Department of Spine SurgeryAffiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Guangjun Jiao
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
| | - Yunhao You
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Xiang Li
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Jincheng Liu
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Zhenqian Sun
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Qinghui Li
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Zihan Dai
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Jinlong Ma
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Hongming Zhou
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Department of Spine SurgeryLinyi Central HospitalLinyiShandongChina
| | - Gang Li
- Department of MicroorthopaedicsAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanShandongChina
| | - Chunyang Meng
- Department of Spine SurgeryAffiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Yunzhen Chen
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
| |
Collapse
|
134
|
Luo HL, Lee YC, Chang YL, Hsu WC, Wu YT, Jhan JH, Lin HH, Wu YR, Ke HL, Liu HY. MicroRNA-145-5p suppresses cell proliferation, migration, and invasion in upper tract urothelial carcinoma by targeting 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase. J Cell Biochem 2023; 124:1324-1345. [PMID: 37475541 DOI: 10.1002/jcb.30449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/22/2023]
Abstract
Upper tract urothelial carcinoma (UTUC), including renal, pelvic, and ureteral carcinoma, has a high incidence rate in Taiwan, which is different from that in Western countries. Therefore, it is imperative to elucidate the mechanisms underlying UTUC growth and metastasis. To explore the function of miR-145-5p in UTUC, we transfected the BFTC909 cell line with miR-145-5p mimics and analyzed the differences in protein levels by performing two-dimensional polyacrylamide gel electrophoresis. Real-time polymerase chain reaction and Western blot analysis were used to analyze 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inositol monophosphate cyclohydrolase (ATIC) messenger RNA and protein levels. A dual-luciferase assay was performed to identify the target of miR-145-5p in ATIC. The effects of miR-145-5p and ATIC expression by cell transfection on cell proliferation, migration, and invasion were also assessed. miR-145-5p downregulated ATIC protein expression. High ATIC expression is associated with tumor stage, metastasis, recurrence, and a poor prognosis in patients with UTUC. Cell function assays revealed that ATIC knockdown inhibited the proliferation, migration, and invasive abilities of UTUC cells. In contrast, miR-145-5p affected the proliferation, migration, and invasive abilities of UTUC cells by directly targeting the 3'-untranslated regions of ATIC. Furthermore, we used RNA sequencing and Ingenuity Pathway Analysis to identify possible downstream genes regulated by ATIC and found that miR-145-5p regulated the protein levels of fibronectin 1, Slug, cyclin A2, cyclin B1, P57, and interferon-induced transmembrane 1 via ATIC. ATIC may be a valuable predictor of prognosis and a potential therapeutic target for UTUC.
Collapse
Affiliation(s)
- Hao-Lun Luo
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University and College of Medicine, Kaohsiung, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Chen Lee
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yin-Lun Chang
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University and College of Medicine, Kaohsiung, Taiwan
| | - Wei-Chi Hsu
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Ting Wu
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University and College of Medicine, Kaohsiung, Taiwan
| | - Jhen-Hao Jhan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
| | - Hui-Hui Lin
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yi-Ru Wu
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hung-Lung Ke
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Hui-Ying Liu
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University and College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
135
|
Armour SL, Stanley JE, Cantley J, Dean ED, Knudsen JG. Metabolic regulation of glucagon secretion. J Endocrinol 2023; 259:e230081. [PMID: 37523232 PMCID: PMC10681275 DOI: 10.1530/joe-23-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/31/2023] [Indexed: 08/01/2023]
Abstract
Since the discovery of glucagon 100 years ago, the hormone and the pancreatic islet alpha cells that produce it have remained enigmatic relative to insulin-producing beta cells. Canonically, alpha cells have been described in the context of glucagon's role in glucose metabolism in liver, with glucose as the primary nutrient signal regulating alpha cell function. However, current data reveal a more holistic model of metabolic signalling, involving glucagon-regulated metabolism of multiple nutrients by the liver and other tissues, including amino acids and lipids, providing reciprocal feedback to regulate glucagon secretion and even alpha cell mass. Here we describe how various nutrients are sensed, transported and metabolised in alpha cells, providing an integrative model for the metabolic regulation of glucagon secretion and action. Importantly, we discuss where these nutrient-sensing pathways intersect to regulate alpha cell function and highlight key areas for future research.
Collapse
Affiliation(s)
- Sarah L Armour
- Section for cell biology and physiology, Department of Biology, University of Copenhagen, DK
| | - Jade E. Stanley
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, USA
| | - James Cantley
- Division of Cellular and systems medicine, School of Medicine, University of Dundee, UK
| | - E. Danielle Dean
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, USA
- Division of Diabetes, Endocrinology, & Metabolism, Vanderbilt University Medical Center school of medicine, USA
| | - Jakob G Knudsen
- Section for cell biology and physiology, Department of Biology, University of Copenhagen, DK
| |
Collapse
|
136
|
Luo X, Wei M, Li W, Zhao H, Kasim V, Wu S. PBX3 promotes pentose phosphate pathway and colorectal cancer progression by enhancing G6PD expression. Int J Biol Sci 2023; 19:4525-4538. [PMID: 37781025 PMCID: PMC10535713 DOI: 10.7150/ijbs.86279] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/19/2023] [Indexed: 10/03/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancers crucial for fulfilling the needs of energy, building blocks, and antioxidants to support tumor cells' rapid proliferation and to cope with the harsh microenvironment. Pre-B-cell leukemia transcription factor 3 (PBX3) is a member of the PBX family whose expression is up-regulated in various tumors, however, whether it is involved in tumor cell metabolic reprogramming remains unclear. Herein, we report that PBX3 is a positive regulator of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme in the pentose phosphate pathway (PPP). PBX3 promoted G6PD transcriptional activity in tumor cells by binding directly to its promoter, leading to PPP stimulation and enhancing the production of nucleotides and NADPH, a crucial reductant, thereby promoting nucleic acid and lipid biosynthesis while decreasing intracellular reactive oxygen species levels. The PBX3/G6PD axis also promoted tumorigenic potential in vitro and in vivo. Collectively, these findings reveal a novel function of PBX3 as a regulator of G6PD, linking its oncogenic activity with tumor cell metabolic reprogramming, especially PPP. Furthermore, our results suggested that PBX3 is a potential target for metabolic-based anti-tumor therapeutic strategies.
Collapse
Affiliation(s)
- Xinxin Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Mankun Wei
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wenfang Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Hezhao Zhao
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China
| |
Collapse
|
137
|
Liu B, Fu X, Du Y, Feng Z, Chen R, Liu X, Yu F, Zhou G, Ba Y. Pan-cancer analysis of G6PD carcinogenesis in human tumors. Carcinogenesis 2023; 44:525-534. [PMID: 37335542 DOI: 10.1093/carcin/bgad043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/24/2023] [Accepted: 06/18/2023] [Indexed: 06/21/2023] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is involved in the catalytic pentose phosphate pathway (PPP), which is closely related to energy metabolism. G6PD plays a crucial role in many types of cancer, but the specific molecular mechanisms of G6PD in cancer remain unclear. Therefore, we investigated the potential oncogenic role of G6PD in various tumors based on The Cancer Genome Atlas (TCGA), the cBioPortal datasets, the University of California Santa Cruz (UCSC) Xena browser, and the UALCAN-based online tool. G6PD was highly expressed in several cancer tissues (hepatocellular carcinoma, glioma, and breast cancer) compared with normal tissues and was significantly associated with poor prognosis of hepatocellular carcinoma, clear cell renal cell carcinoma, and breast cancer. Promoter methylation levels of G6PD were lower in Bladder Urothelial Carcinoma (BLCA) (P = 2.77e-02), breast invasive carcinoma (BRCA) (P = 1.62e-12), kidney renal clear cell carcinoma (KIRC) (P = 4.23e-02), kidney renal papillary cell carcinoma (KIRP) (P = 2.64e-03), liver hepatocellular carcinoma (LIHC) (P = 1.76e-02), stomach adenocarcinoma (STAD) (P = 3.50e-02), testicular germ cell tumors (TGCT) (P = 1.62e-12), higher in prostate adenocarcinoma (PRAD) (P = 1.81e-09), and uterine corpus endometrial carcinoma (UCEC) (P = 2.96e-04) compared with corresponding normal tissue samples. G6PD expression was positively correlated with the infiltration level of immune cells in most tumors, suggesting that G6PD may be involved in tumor immune infiltration. In addition, the functional mechanism of G6PD also involves 'Carbon metabolism', 'Glycolysis/Gluconeogenesis', 'Pentose phosphate pathway', and 'Central carbon pathway metabolism in cancer signaling pathway'. This pan-cancer study provides a relatively broad understanding of the oncogenic role of G6PD in various tumors and presents a theoretical basis for the development of G6PD inhibitors as therapeutic drugs for multiple cancers.
Collapse
Affiliation(s)
- Bin Liu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Xiaoli Fu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Yuhui Du
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Zichen Feng
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Ruiqin Chen
- Jinshui District Center for Disease Control and Prevention, Zhengzhou, Henan 450053, P. R. China
| | - Xiaoxue Liu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Fangfang Yu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Guoyu Zhou
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Yue Ba
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| |
Collapse
|
138
|
Nekooie Marnany N, Fodil R, Féréol S, Dady A, Depp M, Relaix F, Motterlini R, Foresti R, Duband JL, Dufour S. Glucose oxidation drives trunk neural crest cell development and fate. J Cell Sci 2023; 136:jcs260607. [PMID: 37589341 DOI: 10.1242/jcs.260607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 06/30/2023] [Indexed: 08/18/2023] Open
Abstract
Bioenergetic metabolism is a key regulator of cellular function and signaling, but how it can instruct the behavior of cells and their fate during embryonic development remains largely unknown. Here, we investigated the role of glucose metabolism in the development of avian trunk neural crest cells (NCCs), a migratory stem cell population of the vertebrate embryo. We uncovered that trunk NCCs display glucose oxidation as a prominent metabolic phenotype, in contrast to what is seen for cranial NCCs, which instead rely on aerobic glycolysis. In addition, only one pathway downstream of glucose uptake is not sufficient for trunk NCC development. Indeed, glycolysis, mitochondrial respiration and the pentose phosphate pathway are all mobilized and integrated for the coordinated execution of diverse cellular programs, epithelial-to-mesenchymal transition, adhesion, locomotion, proliferation and differentiation, through regulation of specific gene expression. In the absence of glucose, the OXPHOS pathway fueled by pyruvate failed to promote trunk NCC adaptation to environmental stiffness, stemness maintenance and fate-decision making. These findings highlight the need for trunk NCCs to make the most of the glucose pathway potential to meet the high metabolic demands appropriate for their development.
Collapse
Affiliation(s)
| | - Redouane Fodil
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Sophie Féréol
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Alwyn Dady
- Laboratoire Gly-CRRET, Université Paris-Est Créteil, 94000 Créteil, France
| | - Marine Depp
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Frederic Relaix
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | | | - Roberta Foresti
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Jean-Loup Duband
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Sylvie Dufour
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| |
Collapse
|
139
|
Heiserman JP, Minhas Z, Nikpayam E, Cheon DJ. Targeting Heat Shock Protein 27 and Fatty Acid Oxidation Augments Cisplatin Treatment in Cisplatin-Resistant Ovarian Cancer Cell Lines. Int J Mol Sci 2023; 24:12638. [PMID: 37628819 PMCID: PMC10454186 DOI: 10.3390/ijms241612638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Most ovarian cancer patients develop recurrent cancers which are often resistant to commonly employed chemotherapy agents, such as cisplatin. We have previously shown that the inhibition of heat shock protein 27 (HSP27) or fatty acid oxidation (FAO) sensitizes cisplatin-resistant ovarian cancer cell lines to cisplatin and dual inhibition of both HSP27 and FAO induces substantial cell death in vitro. However, it is unclear how HSP27 and FAO promote cisplatin resistance, and if dual inhibition of both HSP27 and FAO would augment cisplatin treatment in vivo. Here we showed that HSP27 knockdown in two cisplatin-resistant ovarian cancer cell lines (A2780CIS and PEO4) resulted in more ROS production upon cisplatin treatment. HSP27-knockdown cancer cells exhibited decreased levels of reduced glutathione (GSH) and glucose6phosphate dehydrogenase (G6PD), a crucial pentose phosphate pathway enzyme. ROS depletion with the compound N-acetyl cysteine (NAC) attenuated cisplatin-induced upregulation of HSP27, FAO, and markers of apoptosis and ferroptosis in cisplatin-resistant ovarian cancer cell lines. Finally, inhibition of HSP27 and FAO with ivermectin and perhexiline enhanced the cytotoxic effect of cisplatin in A2780CIS xenograft tumors in vivo. Our results suggest that two different cisplatin-resistant ovarian cancer cell lines upregulate HSP27 and FAO to deplete cisplatin-induced ROS to attenuate cisplatin's cytotoxic effect.
Collapse
Affiliation(s)
| | | | | | - Dong-Joo Cheon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA; (J.P.H.); (E.N.)
| |
Collapse
|
140
|
Gallego-López GM, Guzman EC, Knoll LJ, Skala M. Metabolic changes to host cells with Toxoplasma gondii infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552811. [PMID: 37609172 PMCID: PMC10441426 DOI: 10.1101/2023.08.10.552811] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Toxoplasma gondii, the causative agent of toxoplasmosis, is an obligate intracellular parasite that infects warm-blooded vertebrates across the world. In humans, seropositivity rates of T. gondii range from 10% to 90%. Despite its prevalence, few studies address how T. gondii infection changes the metabolism of host cells. Here, we investigate how T. gondii manipulates the host cell metabolic environment by monitoring metabolic response over time using non-invasive autofluorescence lifetime imaging of single cells, seahorse metabolic flux analysis, reactive oxygen species (ROS) production, and metabolomics. Autofluorescence lifetime imaging indicates that infected host cells become more oxidized and have an increased proportion of bound NAD(P)H with infection. These findings are consistent with changes in mitochondrial and glycolytic function, decrease of intracellular glucose, fluctuations in lactate and ROS production in infected cells over time. We also examined changes associated with the pre-invasion "kiss and spit" process using autofluorescence lifetime imaging, which similarly showed a more oxidized host cell with an increased proportion of bound NAD(P)H over 48 hours. Glucose metabolic flux analysis indicated that these changes are driven by NADH and NADP+ in T. gondii infection. In sum, metabolic changes in host cells with T. gondii infection were similar during full infection, and kiss and spit. Autofluorescence lifetime imaging can non-invasively monitor metabolic changes in host cells over a microbial infection time-course.
Collapse
Affiliation(s)
- Gina M. Gallego-López
- Morgridge Institute for Research, Madison, WI, 53706
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, WI, 53706
| | | | - Laura J. Knoll
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, WI, 53706
| | - Melissa Skala
- Morgridge Institute for Research, Madison, WI, 53706
- Department of Biomedical Engineering, University of Wisconsin- Madison, WI 53706, USA
| |
Collapse
|
141
|
Ndlovu IS, Tshilwane SI, Vosloo A, Chaisi M, Mukaratirwa S. Metabolomics of Type 2 Diabetes Mellitus in Sprague Dawley Rats-In Search of Potential Metabolic Biomarkers. Int J Mol Sci 2023; 24:12467. [PMID: 37569840 PMCID: PMC10419637 DOI: 10.3390/ijms241512467] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is an expanding global health concern, closely associated with the epidemic of obesity. Individuals with diabetes are at high risk for microvascular and macrovascular complications, which include retinopathy, neuropathy, and cardiovascular comorbidities. Despite the availability of diagnostic tools for T2DM, approximately 30-60% of people with T2DM in developed countries are never diagnosed or detected. Therefore, there is a strong need for a simpler and more reliable technique for the early detection of T2DM. This study aimed to use a non-targeted metabolomic approach to systematically identify novel biomarkers from the serum samples of T2DM-induced Sprague Dawley (SD) rats using a comprehensive two-dimensional gas chromatography coupled with a time-of-flight mass spectrometry (GCxGC-TOF/MS). Fifty-four male Sprague Dawley rats weighing between 160-180 g were randomly assigned into two experimental groups, namely the type 2 diabetes mellitus group (T2DM) (n = 36) and the non-diabetic control group (n = 18). Results from this study showed that the metabolite signature of the diabetic rats was different from that of the non-diabetic control group. The most significantly upregulated metabolic pathway was aminoacyl-t-RNA biosynthesis. Metabolite changes observed between the diabetic and non-diabetic control group was attributed to the increase in amino acids, such as glycine, L-asparagine, and L-serine. Aromatic amino acids, including L-tyrosine, were associated with the risk of future hyperglycemia and overt diabetes. The identified potential biomarkers depicted a good predictive value of more than 0.8. It was concluded from the results that amino acids that were associated with impaired insulin secretion were prospectively related to an increase in glucose levels. Moreover, amino acids that were associated with impaired insulin secretion were prospectively related to an increase in glucose levels.
Collapse
Affiliation(s)
- Innocent Siyanda Ndlovu
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa; (I.S.N.); (A.V.)
| | - Selaelo Ivy Tshilwane
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria 0110, South Africa; (S.I.T.); (M.C.)
| | - Andre Vosloo
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa; (I.S.N.); (A.V.)
| | - Mamohale Chaisi
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria 0110, South Africa; (S.I.T.); (M.C.)
- Foundational Biodiversity Science, South African National Biodiversity Institute, Pretoria 0001, South Africa
| | - Samson Mukaratirwa
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa; (I.S.N.); (A.V.)
- One Health Center for Zoonoses and Tropical Veterinary Medicine, School of Veterinary Medicine, Ross University, Basseterre KN0101, Saint Kitts and Nevis
| |
Collapse
|
142
|
Muniz-Santos R, Lucieri-Costa G, de Almeida MAP, Moraes-de-Souza I, Brito MADSM, Silva AR, Gonçalves-de-Albuquerque CF. Lipid oxidation dysregulation: an emerging player in the pathophysiology of sepsis. Front Immunol 2023; 14:1224335. [PMID: 37600769 PMCID: PMC10435884 DOI: 10.3389/fimmu.2023.1224335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/30/2023] [Indexed: 08/22/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by abnormal host response to infection. Millions of people are affected annually worldwide. Derangement of the inflammatory response is crucial in sepsis pathogenesis. However, metabolic, coagulation, and thermoregulatory alterations also occur in patients with sepsis. Fatty acid mobilization and oxidation changes may assume the role of a protagonist in sepsis pathogenesis. Lipid oxidation and free fatty acids (FFAs) are potentially valuable markers for sepsis diagnosis and prognosis. Herein, we discuss inflammatory and metabolic dysfunction during sepsis, focusing on fatty acid oxidation (FAO) alterations in the liver and muscle (skeletal and cardiac) and their implications in sepsis development.
Collapse
Affiliation(s)
- Renan Muniz-Santos
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giovanna Lucieri-Costa
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus Augusto P. de Almeida
- Neuroscience Graduate Program, Federal Fluminense University, Niteroi, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Isabelle Moraes-de-Souza
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Adriana Ribeiro Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroscience Graduate Program, Federal Fluminense University, Niteroi, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
143
|
Zeng T, Li B, Shu X, Pang J, Wang H, Cai X, Liao Y, Xiao X, Chong Y, Gong J, Li X. Pan-cancer analysis reveals that G6PD is a prognostic biomarker and therapeutic target for a variety of cancers. Front Oncol 2023; 13:1183474. [PMID: 37601657 PMCID: PMC10435888 DOI: 10.3389/fonc.2023.1183474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Background Despite accumulating evidence revealing that Glucose-6-phosphate dehydrogenase (G6PD) is highly expressed in many tumor tissues and plays a remarkable role in cancer tumorigenesis and progression, there is still a lack of G6PD pan-cancer analysis. This study was designed to analyze the expression status and prognostic significance of G6PD in pan-cancer. Methods G6PD expression data were obtained from multiple data resources including the Genotype-Tissue Expression, the Cancer Genome Atlas, and the Tumor Immunity Estimation Resource. These data were used to assess the G6PD expression, prognostic value, and clinical characteristics. The ESTIMATE algorithms were used to analyze the association between G6PD expression and immune-infiltrating cells and the tumor microenvironment. The functional enrichment analysis was also performed across pan-cancer. In addition, the GDSC1 database containing 403 drugs was utilized to explore the relationship between drug sensitivity and G6PD expression levels. Furthermore, we also performed clinical validation and in vitro experiments to further validate the role of G6PD in hepatocellular carcinoma (HCC) cells and its correlation with prognosis. The R software was used for statistical analysis and data visualization. Results G6PD expression was upregulated in most cancers compared to their normal counterparts. The study also revealed that G6PD expression was a prognostic indicator and high levels of G6PD expression were correlated with worse clinical prognosis including overall survival, disease-specific survival, and progression-free interval in multiple cancers. Furthermore, the G6PD level was also related to cancer immunity infiltration in most of the cancers, especially in KIRC, LGG, and LIHC. In addition to this, G6PD expression was positively related to pathological stages of KIRP, BRCA, KIRC, and LIHC. Functional analysis and protein-protein interactions network results revealed that G6PD was involved in metabolism-related activities, immune responses, proliferation, and apoptosis. Drug sensitivity analysis showed that IC50 values of most identified anti-cancer drugs were positively correlated with the G6PD expression. Notably, in vitro functional validation showed that G6PD knockdown attenuated the phenotypes of proliferation in HCC. Conclusion G6PD may serve as a potential prognostic biomarker for cancers and may be a potential therapeutic target gene for tumor therapy.
Collapse
Affiliation(s)
- Tao Zeng
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Infectious Diseases, The First People’s Hospital of Kashi Prefecture, Kashi, China
| | - Bin Li
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xin Shu
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jiahui Pang
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Heping Wang
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xianghao Cai
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yingying Liao
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaolong Xiao
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yutian Chong
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jiao Gong
- Department of Laboratory Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xinhua Li
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
144
|
Jiang X, Peng J, Xie Y, Xu Y, Liu Q, Cheng C, Yan P, Xu S, Wang Y, Zhang L, Li H, Li Y, Li B, Han J, Yu D. Oxoglutarate dehydrogenase-like inhibits the progression of hepatocellular carcinoma by inducing DNA damage through non-canonical function. Cell Death Differ 2023; 30:1931-1942. [PMID: 37419985 PMCID: PMC10406884 DOI: 10.1038/s41418-023-01186-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/09/2023] Open
Abstract
Oxoglutarate dehydrogenase-like (OGDHL) is considered to be the isoenzyme of oxyglutarate dehydrogenase (OGDH) in the OGDH complex, which degrades glucose and glutamate. OGDHL was reported to reprogram glutamine metabolism to suppress HCC progression in an enzyme-activity-dependent manner. However, the potential subcellular localization and non-canonical function of OGDHL is poorly understood. We investigated the expression of OGDHL and its effect on HCC progression. By employing a variety of molecular biology techniques, we revealed the underlying mechanism of OGDHL-induced DNA damage in HCC cells in vitro and in vivo. AAV loaded with OGDHL exerts therapeutic effect on mouse HCC and prolongs survival time. OGDHL induces DNA damage in HCC cells in vitro and in vivo. We also observed that OGDHL possesses nuclear localization in HCC cells and OGDHL-induced DNA damage was independent of its enzymatic activity. Mechanistically, it was demonstrated that OGDHL binds to CDK4 in the nucleus to inhibit the phosphorylation of CDK4 by CAK, which in turn attenuates E2F1 signaling. Inhibition of E2F1 signaling downregulates pyrimidine and purine synthesis, thereby inducing DNA damage through dNTP depletion. We clarified the nuclear localization of OGDHL and its non-canonical function to induce DNA damage, which demonstrated that OGDHL may serve as a select potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Xiang Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Jin Peng
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Yuanyuan Xie
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Yanchao Xu
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, 210008, China
| | - Qi Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Chunxiao Cheng
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Peng Yan
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Shoujing Xu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, China
| | - Ye Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Laizhu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Huan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Yunzheng Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Binghua Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Junhai Han
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, China
| | - Decai Yu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China.
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, 210008, China.
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| |
Collapse
|
145
|
Catalano G, Zaza A, Banella C, Pelosi E, Castelli G, de Marinis E, Smigliani A, Travaglini S, Ottone T, Divona M, Del Principe MI, Buccisano F, Maurillo L, Ammatuna E, Testa U, Nervi C, Venditti A, Voso MT, Noguera NI. MCL1 regulates AML cells metabolism via direct interaction with HK2. Metabolic signature at onset predicts overall survival in AMLs' patients. Leukemia 2023; 37:1600-1610. [PMID: 37349598 DOI: 10.1038/s41375-023-01946-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/31/2023] [Accepted: 06/14/2023] [Indexed: 06/24/2023]
Abstract
We characterize the metabolic background in distinct Acute Myeloid Leukemias (AMLs), by comparing the metabolism of primary AML blasts isolated at diagnosis with that of normal hematopoietic maturing progenitors, using the Seahorse XF Agilent. Leukemic cells feature lower spare respiratory (SRC) and glycolytic capacities as compared to hematopoietic precursors (i.e. day 7, promyelocytes). According with Proton Leak (PL) values, AML blasts can be grouped in two well defined populations. The AML group with blasts presenting high PL or high basal OXPHOS plus high SRC levels had shorter overall survival time and significantly overexpressed myeloid cell leukemia 1 (MCL1) protein. We demonstrate that MCL1 directly binds to Hexokinase 2 (HK2) on the outer mitochondrial membrane (OMM). Overall, these results suggest that high PL and high SRC plus high basal OXPHOS levels at disease onset, arguably with the concourse of MCL1/HK2 action, are significantly linked with shorter overall survival time in AML. Our data describe a new function for MCL1 protein in AMLs' cells: by forming a complex with HK2, MCL1 co-localizes to VDAC on the OMM, thus inducing glycolysis and OXPHOS, ultimately conferring metabolic plasticity and promoting resistance to therapy.
Collapse
Affiliation(s)
- Gianfranco Catalano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
| | - Alessandra Zaza
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
| | - Cristina Banella
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, Florence, Italy
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elisabetta de Marinis
- Department of Medical and Surgical Sciences and Biotechnologies, University of Roma La Sapienza, Rome, Italy
| | - Ariela Smigliani
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
| | - Serena Travaglini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy
| | - Mariadomenica Divona
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Francesco Buccisano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Luca Maurillo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Emanuele Ammatuna
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Clara Nervi
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Adriano Venditti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy.
| | - Nelida Ines Noguera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, Rome, Italy.
| |
Collapse
|
146
|
Mohamed Abdoul-Latif F, Ainane A, Houmed Aboubaker I, Mohamed J, Ainane T. Exploring the Potent Anticancer Activity of Essential Oils and Their Bioactive Compounds: Mechanisms and Prospects for Future Cancer Therapy. Pharmaceuticals (Basel) 2023; 16:1086. [PMID: 37631000 PMCID: PMC10458506 DOI: 10.3390/ph16081086] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide, affecting millions of people each year. Fortunately, the last decades have been marked by considerable advances in the field of cancer therapy. Researchers have discovered many natural substances, some of which are isolated from plants that have promising anti-tumor activity. Among these, essential oils (EOs) and their constituents have been widely studied and shown potent anticancer activities, both in vitro and in vivo. However, despite the promising results, the precise mechanisms of action of EOs and their bioactive compounds are still poorly understood. Further research is needed to better understand these mechanisms, as well as their effectiveness and safety in use. Furthermore, the use of EOs as anticancer drugs is complex, as it requires absolute pharmacodynamic specificity and selectivity, as well as an appropriate formulation for effective administration. In this study, we present a synthesis of recent work on the mechanisms of anticancer action of EOs and their bioactive compounds, examining the results of various in vitro and in vivo studies. We also review future research prospects in this exciting field, as well as potential implications for the development of new cancer drugs.
Collapse
Affiliation(s)
- Fatouma Mohamed Abdoul-Latif
- Medicinal Research Institute, Center for Studies and Research of Djibouti, IRM-CERD, Route de l’Aéroport, Haramous, Djibouti City P.O. Box 486, Djibouti;
| | - Ayoub Ainane
- Superior School of Technology of Khenifra, University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco; (A.A.); (T.A.)
| | | | - Jalludin Mohamed
- Medicinal Research Institute, Center for Studies and Research of Djibouti, IRM-CERD, Route de l’Aéroport, Haramous, Djibouti City P.O. Box 486, Djibouti;
| | - Tarik Ainane
- Superior School of Technology of Khenifra, University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco; (A.A.); (T.A.)
| |
Collapse
|
147
|
Ji S, Feng L, Fu Z, Wu G, Wu Y, Lin Y, Lu D, Song Y, Cui P, Yang Z, Sang C, Song G, Cai S, Li Y, Lin H, Zhang S, Wang X, Qiu S, Zhang X, Hua G, Li J, Zhou J, Dai Z, Wang X, Ding L, Wang P, Gao D, Zhang B, Rodriguez H, Fan J, Clevers H, Zhou H, Sun Y, Gao Q. Pharmaco-proteogenomic characterization of liver cancer organoids for precision oncology. Sci Transl Med 2023; 15:eadg3358. [PMID: 37494474 PMCID: PMC10949980 DOI: 10.1126/scitranslmed.adg3358] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/22/2023] [Indexed: 07/28/2023]
Abstract
Organoid models have the potential to recapitulate the biological and pharmacotypic features of parental tumors. Nevertheless, integrative pharmaco-proteogenomics analysis for drug response features and biomarker investigation for precision therapy of patients with liver cancer are still lacking. We established a patient-derived liver cancer organoid biobank (LICOB) that comprehensively represents the histological and molecular characteristics of various liver cancer types as determined by multiomics profiling, including genomic, epigenomic, transcriptomic, and proteomic analysis. Proteogenomic profiling of LICOB identified proliferative and metabolic organoid subtypes linked to patient prognosis. High-throughput drug screening revealed distinct response patterns of each subtype that were associated with specific multiomics signatures. Through integrative analyses of LICOB pharmaco-proteogenomics data, we identified the molecular features associated with drug responses and predicted potential drug combinations for personalized patient treatment. The synergistic inhibition effect of mTOR inhibitor temsirolimus and the multitargeted tyrosine kinase inhibitor lenvatinib was validated in organoids and patient-derived xenografts models. We also provide a user-friendly web portal to help serve the biomedical research community. Our study is a rich resource for investigation of liver cancer biology and pharmacological dependencies and may help enable functional precision medicine.
Collapse
Affiliation(s)
- Shuyi Ji
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai 201508, China
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Li Feng
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zile Fu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Gaohua Wu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Yingcheng Wu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Youpei Lin
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Dayun Lu
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuanli Song
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Peng Cui
- Burning Rock Biotech, Shanghai 201114, China
| | - Zijian Yang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Chen Sang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Guohe Song
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Shangli Cai
- Burning Rock Biotech, Shanghai 201114, China
| | | | - Hanqing Lin
- D1 Medical Technology, Shanghai 200235, China
| | - Shu Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Xiaoying Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Shuangjian Qiu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Xiaoming Zhang
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guoqiang Hua
- Department of Radiation Oncology, and Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Junqiang Li
- D1 Medical Technology, Shanghai 200235, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zhi Dai
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital Institute for Clinical Science, Fudan University Shanghai Medical College, Shanghai, China
| | - Li Ding
- Department of Medicine, McDonnell Genome Institute, Siteman Cancer Center, Washington University, St. Louis, MO 63108, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NewYork, NY 10029, USA
| | - Daming Gao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
- Department of Radiation Oncology, and Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands
- Current Address: Roche Pharma Research and Early Development (pRED), Basel, Switzerland
| | - Hu Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute of Materia Medica-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology
| | - Yidi Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiang Gao
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai 201508, China
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai 200433, China
| |
Collapse
|
148
|
Kang M, Kang JH, Sim IA, Seong DY, Han S, Jang H, Lee H, Kang SW, Kim SY. Glucose Deprivation Induces Cancer Cell Death through Failure of ROS Regulation. Int J Mol Sci 2023; 24:11969. [PMID: 37569345 PMCID: PMC10418724 DOI: 10.3390/ijms241511969] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
In previous work, we showed that cancer cells do not depend on glycolysis for ATP production, but they do on fatty acid oxidation. However, we found some cancer cells induced cell death after glucose deprivation along with a decrease of ATP production. We investigated the different response of glucose deprivation with two types of cancer cells including glucose insensitive cancer cells (GIC) which do not change ATP levels, and glucose sensitive cancer cells (GSC) which decrease ATP production in 24 h. Glucose deprivation-induced cell death in GSC by more than twofold after 12 h and by up to tenfold after 24 h accompanied by decreased ATP production to compare to the control (cultured in glucose). Glucose deprivation decreased the levels of metabolic intermediates of the pentose phosphate pathway (PPP) and the reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) in both GSC and GIC. However, glucose deprivation increased reactive oxygen species (ROS) only in GSC, suggesting that GIC have a higher tolerance for decreased NADPH than GSC. The twofold higher ratio of reduced/oxidized glutathione (GSH/GSSG) in GIS than in GSC correlates closely with the twofold lower ROS levels under glucose starvation conditions. Treatment with N-acetylcysteine (NAC) as a precursor to the biologic antioxidant glutathione restored ATP production by 70% and reversed cell death caused by glucose deprivation in GSC. The present findings suggest that glucose deprivation-induced cancer cell death is not caused by decreased ATP levels, but rather triggered by a failure of ROS regulation by the antioxidant system. Conclusion is clear that glucose deprivation-induced cell death is independent from ATP depletion-induced cell death.
Collapse
Affiliation(s)
- Mingyu Kang
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Gyeonggi-do, Republic of Korea; (M.K.); (J.H.K.); (I.A.S.); (D.Y.S.); (H.L.)
- New Cancer Cure Bio Co., Goyang 10408, Gyeonggi-do, Republic of Korea
| | - Joon H. Kang
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Gyeonggi-do, Republic of Korea; (M.K.); (J.H.K.); (I.A.S.); (D.Y.S.); (H.L.)
- New Cancer Cure Bio Co., Goyang 10408, Gyeonggi-do, Republic of Korea
| | - In A. Sim
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Gyeonggi-do, Republic of Korea; (M.K.); (J.H.K.); (I.A.S.); (D.Y.S.); (H.L.)
- New Cancer Cure Bio Co., Goyang 10408, Gyeonggi-do, Republic of Korea
| | - Do Y. Seong
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Gyeonggi-do, Republic of Korea; (M.K.); (J.H.K.); (I.A.S.); (D.Y.S.); (H.L.)
| | - Suji Han
- Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang 10408, Gyeonggi-do, Republic of Korea; (S.H.); (H.J.)
| | - Hyonchol Jang
- Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang 10408, Gyeonggi-do, Republic of Korea; (S.H.); (H.J.)
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Gyeonggi-do, Republic of Korea
| | - Ho Lee
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Gyeonggi-do, Republic of Korea; (M.K.); (J.H.K.); (I.A.S.); (D.Y.S.); (H.L.)
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Gyeonggi-do, Republic of Korea
| | - Sang W. Kang
- Department of Life Science, Ewha Women’s University, Seoul 03760, Republic of Korea;
| | - Soo-Youl Kim
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Gyeonggi-do, Republic of Korea; (M.K.); (J.H.K.); (I.A.S.); (D.Y.S.); (H.L.)
- New Cancer Cure Bio Co., Goyang 10408, Gyeonggi-do, Republic of Korea
| |
Collapse
|
149
|
Li M, Yang Y, Xiong L, Jiang P, Wang J, Li C. Metabolism, metabolites, and macrophages in cancer. J Hematol Oncol 2023; 16:80. [PMID: 37491279 PMCID: PMC10367370 DOI: 10.1186/s13045-023-01478-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/13/2023] [Indexed: 07/27/2023] Open
Abstract
Tumour-associated macrophages (TAMs) are crucial components of the tumour microenvironment and play a significant role in tumour development and drug resistance by creating an immunosuppressive microenvironment. Macrophages are essential components of both the innate and adaptive immune systems and contribute to pathogen resistance and the regulation of organism homeostasis. Macrophage function and polarization are closely linked to altered metabolism. Generally, M1 macrophages rely primarily on aerobic glycolysis, whereas M2 macrophages depend on oxidative metabolism. Metabolic studies have revealed that the metabolic signature of TAMs and metabolites in the tumour microenvironment regulate the function and polarization of TAMs. However, the precise effects of metabolic reprogramming on tumours and TAMs remain incompletely understood. In this review, we discuss the impact of metabolic pathways on macrophage function and polarization as well as potential strategies for reprogramming macrophage metabolism in cancer treatment.
Collapse
Affiliation(s)
- Mengyuan Li
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Yuhan Yang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Liting Xiong
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Ping Jiang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.
| | - Chunxiao Li
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
150
|
Domínguez-Zorita S, Cuezva JM. The Mitochondrial ATP Synthase/IF1 Axis in Cancer Progression: Targets for Therapeutic Intervention. Cancers (Basel) 2023; 15:3775. [PMID: 37568591 PMCID: PMC10417293 DOI: 10.3390/cancers15153775] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Cancer poses a significant global health problem with profound personal and economic implications on National Health Care Systems. The reprograming of metabolism is a major trait of the cancer phenotype with a clear potential for developing effective therapeutic strategies to combat the disease. Herein, we summarize the relevant role that the mitochondrial ATP synthase and its physiological inhibitor, ATPase Inhibitory Factor 1 (IF1), play in metabolic reprogramming to an enhanced glycolytic phenotype. We stress that the interplay in the ATP synthase/IF1 axis has additional functional roles in signaling mitohormetic programs, pro-oncogenic or anti-metastatic phenotypes depending on the cell type. Moreover, the same axis also participates in cell death resistance of cancer cells by restrained mitochondrial permeability transition pore opening. We emphasize the relevance of the different post-transcriptional mechanisms that regulate the specific expression and activity of ATP synthase/IF1, to stimulate further investigations in the field because of their potential as future targets to treat cancer. In addition, we review recent findings stressing that mitochondria metabolism is the primary altered target in lung adenocarcinomas and that the ATP synthase/IF1 axis of OXPHOS is included in the most significant signature of metastatic disease. Finally, we stress that targeting mitochondrial OXPHOS in pre-clinical mouse models affords a most effective therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Sonia Domínguez-Zorita
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28041 Madrid, Spain
| | - José M. Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28041 Madrid, Spain
| |
Collapse
|